Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Transplantation ; 104(4): 813-822, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31385929

RESUMO

BACKGROUND: Worldwide, a final crossmatch is the gold standard for determining compatibility between patient and donor before solid organ transplantation and preventing hyperacute rejection. In the absence of autoantibodies, an incompatible crossmatch in a sensitized patient is attributed to mismatched donor HLA. However, current physical crossmatch methods cannot distinguish reactivity to HLA from other clinically irrelevant cell surface targets nor the class of HLA if it is the target. Result interpretation is difficult or impossible when autoantibodies, alloantibodies, or therapeutic antibodies coexist. METHODS: Herein, we describe a unique donor-specific flow crossmatch (DSA-FXM) that distinguishes HLA class I or II donor-specific antibody bound to HLA antigens on the donor cell surface in their native conformation that is not impacted by rituximab, anti-thymocyte globulin (after absorption), or autoantibodies. It is HLA specific. RESULTS: We compared the results of single-antigen antibody testing, autoreactive and alloreactive flow cytometry crossmatches (FXM) using traditional FXM and our DSA-FXM method from 94 patients (enriched for auto+/allo+ pairs; n = 64) against 110 donors (338 tests) and show that, in our cohort, positive traditional FXM results are not directed to donor HLA 60.25% of the time and negative traditional FXM results are missing HLA donor-specific antibody 36.2% of the time based on the DSA-FXM. CONCLUSIONS: We demonstrate that the DSA-FXM is able to define categorically distinct and clinically important HLA antibody profiles in half the time required for the standard FXM, potentially shortening cold ischemia time and providing clinicians with unambiguous essential information regarding HLA compatibility when time is critical.


Assuntos
Soro Antilinfocitário/sangue , Autoanticorpos/sangue , Seleção do Doador , Citometria de Fluxo , Antígenos HLA/imunologia , Teste de Histocompatibilidade , Histocompatibilidade , Isoanticorpos/sangue , Transplante de Órgãos , Rituximab/sangue , Biomarcadores/sangue , Tomada de Decisão Clínica , Reações Falso-Positivas , Antígenos HLA/classificação , Humanos , Transplante de Órgãos/efeitos adversos , Valor Preditivo dos Testes , Reprodutibilidade dos Testes , Fatores de Tempo , Fluxo de Trabalho
2.
Hum Immunol ; 80(9): 668-702, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31122739

RESUMO

Current models describing HLA epitopes are both theoretical and empirical. Each has limitations yielding discordant results and increasingly complex modeling. The models make a priori assumptions that epitopes must be present only on the mature protein, solvent accessible, on the 'top' (peptide binding surface) of the molecule, restricted to the same class as the antibody, and in the same position on the target allele if reactive to more than one locus. Results obtained counter to these assumptions are routinely discounted. For the 17th International Histocompatibility and Immunogenetics Workshop, we developed a reverse engineering algorithm to define epitopes without these assumptions on a cohort of 332 primary transplant pairs. Complete NGS typing of the transcribed (including leader) genomic DNA for 11 HLA loci of donor and recipient and DSA assignment by single antigen beads was performed. Our results show that, when grouped by 16 class I and II allele specific DSA, uniform clusters and 172 specific amino acid target epitopes are recognized by recipients despite originating from disparate HLA pairs. Data also show that these targets can be in the leader, alpha 3, transmembrane and cytoplasmic domains, thus calling into question current assumptions regarding immunogenic epitopes. Comparisons of amino acid epitopes defined by the Terasaki and Duquesnoy groups (TerEp and EpRegistry) are given.


Assuntos
Algoritmos , Mapeamento de Epitopos/métodos , Epitopos/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Teste de Histocompatibilidade/métodos , Alelos , Sequência de Aminoácidos , Estudos de Coortes , Epitopos/química , Loci Gênicos , Transplante de Coração , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imunoglobulina G/genética , Transplante de Rim , Projetos Piloto , Domínios Proteicos/imunologia , Doadores de Tecidos , Transplantados
3.
Hum Immunol ; 80(9): 661-667, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30978444

RESUMO

Central to the idea of antibody recognition is some degree of foreignness of the target antigen compared to the antibody producer. Epitopes are distinct regions on an antigen to which antibody can be elicited and bound. However, for HLA antigens, there is no consensus definition of what represents the minimal functional immunogenic unit of dissimilarity. To assess this in an unbiased way, we developed a reverse engineering software strategy based on donor specific antibodies defined by single antigen beads and full length genomic high resolution HLA typing by NGS of recipients and donors (332 transplant pairs). Starting with the ATG of Exon 1 and moving stepwise one amino acid at a time for each of the following triplets, the algorithm compared every possible amino acid triplet of the recipient and donor for 11 loci (A, B, C, DRB1, DRB3, DRB4, DRB5, DQA1, DQB1, DPA1, DPB1). Results were agnostic with respect to HLA class, not restricted to just the mature protein, and not influenced by existing maps (e.g., IMGT, or epitope models). We also developed web-based functions in the 17th IHIWS database to collect the unbiased triplets so that we could group the transplant pairs with the same donor specific antibodies and find shared triplets within the groups as potential core or essential epitopes that trigger the antibody formation. Profiling the pairs where the same DSA was identified led to identification of discrete amino acid triplets shared among the pairs irrespective of HLA match. The potential epitopes were mapped onto the 3D protein structure for reference.


Assuntos
Algoritmos , Anticorpos/imunologia , Epitopos/imunologia , Genótipo , Antígenos HLA/genética , Sequenciamento de Nucleotídeos em Larga Escala , Teste de Histocompatibilidade/métodos , Software , Alelos , Sequência de Aminoácidos/genética , Anticorpos/genética , Estudos de Coortes , Mapeamento de Epitopos , Epitopos/química , Éxons/genética , Antígenos HLA/imunologia , Transplante de Coração , Humanos , Transplante de Rim , Estrutura Terciária de Proteína , Doadores de Tecidos , Transplantados
4.
Circulation ; 139(12): e553-e578, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30776902

RESUMO

Sensitization, defined as the presence of circulating antibodies, presents challenges for heart transplant recipients and physicians. When present, sensitization can limit a transplantation candidate's access to organs, prolong wait time, and, in some cases, exclude the candidate from heart transplantation altogether. The management of sensitization is not yet standardized, and current therapies have not yielded consistent results. Although current strategies involve antibody suppression and removal with intravenous immunoglobulin, plasmapheresis, and antibody therapy, newer strategies with more specific targets are being investigated.


Assuntos
Rejeição de Enxerto/prevenção & controle , Transplante de Coração , Rejeição de Enxerto/etiologia , Antígenos HLA/imunologia , Transplante de Coração/efeitos adversos , Teste de Histocompatibilidade , Humanos , Imunoglobulinas Intravenosas/uso terapêutico , Isoanticorpos/sangue , Isoanticorpos/imunologia , Troca Plasmática , Plasmaferese , Rituximab/uso terapêutico
6.
PLoS Med ; 15(5): e1002572, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29799874

RESUMO

BACKGROUND: Anti-human leukocyte antigen donor-specific antibodies (anti-HLA DSAs) are recognized as a major barrier to patients' access to organ transplantation and the major cause of graft failure. The capacity of circulating anti-HLA DSAs to activate complement has been suggested as a potential biomarker for optimizing graft allocation and improving the rate of successful transplantations. METHODS AND FINDINGS: To address the clinical relevance of complement-activating anti-HLA DSAs across all solid organ transplant patients, we performed a meta-analysis of their association with transplant outcome through a systematic review, from inception to January 31, 2018. The primary outcome was allograft loss, and the secondary outcome was allograft rejection. A comprehensive search strategy was conducted through several databases (Medline, Embase, Cochrane, and Scopus). A total of 5,861 eligible citations were identified. A total of 37 studies were included in the meta-analysis. Studies reported on 7,936 patients, including kidney (n = 5,991), liver (n = 1,459), heart (n = 370), and lung recipients (n = 116). Solid organ transplant recipients with circulating complement-activating anti-HLA DSAs experienced an increased risk of allograft loss (pooled HR 3.09; 95% CI 2.55-3.74, P = 0.001; I2 = 29.3%), and allograft rejection (pooled HR 3.75; 95% CI: 2.05-6.87, P = 0.001; I2 = 69.8%) compared to patients without complement-activating anti-HLA DSAs. The association between circulating complement-activating anti-HLA DSAs and allograft failure was consistent across all subgroups and sensitivity analyses. Limitations of the study are the observational and retrospective design of almost all included studies, the higher proportion of kidney recipients compared to other solid organ transplant recipients, and the inclusion of fewer studies investigating allograft rejection. CONCLUSIONS: In this study, we found that circulating complement-activating anti-HLA DSAs had a significant deleterious impact on solid organ transplant survival and risk of rejection. The detection of complement-activating anti-HLA DSAs may add value at an individual patient level for noninvasive biomarker-guided risk stratification. TRIAL REGISTRATION: National Clinical Trial protocol ID: NCT03438058.


Assuntos
Anticorpos/imunologia , Ativação do Complemento/imunologia , Sobrevivência de Enxerto/imunologia , Antígenos HLA/imunologia , Imunologia de Transplantes/imunologia , Rejeição de Enxerto/imunologia , Humanos
7.
Am J Transplant ; 18(9): 2285-2294, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29687961

RESUMO

Donor-specific antibodies (DSA) to mismatched human leukocyte antigens (HLA) are associated with worse outcomes after lung transplantation. To determine the incidence and characteristics of DSA early after lung transplantation, we conducted a prospective multicenter observational study that used standardized treatment and testing protocols. Among 119 transplant recipients, 43 (36%) developed DSA: 6 (14%) developed DSA only to class I HLA, 23 (53%) developed DSA only to class II HLA, and 14 (33%) developed DSA to both class I and class II HLA. The median DSA mean fluorescence intensity (MFI) was 3197. We identified a significant association between the Lung Allocation Score and the development of DSA (HR = 1.02, 95% CI: 1.001-1.03, P = .047) and a significant association between DSA with an MFI ≥ 3000 and acute cellular rejection (ACR) grade ≥ A2 (HR = 2.11, 95% CI: 1.04-4.27, P = .039). However, we did not detect an association between DSA and survival. We conclude that DSA occur frequently early after lung transplantation, and most target class II HLA. DSA with an MFI ≥ 3000 have a significant association with ACR. Extended follow-up is necessary to determine the impact of DSA on other important outcomes.


Assuntos
Rejeição de Enxerto/mortalidade , Sobrevivência de Enxerto/imunologia , Antígenos HLA/imunologia , Isoanticorpos/efeitos adversos , Transplante de Pulmão/mortalidade , Doadores de Tecidos , Adulto , Idoso , Feminino , Seguimentos , Rejeição de Enxerto/etiologia , Rejeição de Enxerto/patologia , Teste de Histocompatibilidade , Humanos , Isoanticorpos/imunologia , Transplante de Pulmão/efeitos adversos , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias , Prognóstico , Estudos Prospectivos , Fatores de Risco , Taxa de Sobrevida
8.
Curr Opin Organ Transplant ; 22(5): 505-510, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28723698

RESUMO

PURPOSE OF REVIEW: Significant interest and controversy surround the use of C1q for determining risk of antibody-mediated rejection (AMR) and graft loss. Alternate models for predicting outcomes have been proposed. This review focuses on the correlation of currently utilized assays for outcome, together with the technical and theoretical limitations, to distill current thinking. RECENT FINDINGS: Results demonstrate that C1q status is significantly correlated with AMR and graft loss. There is general consensus that C1q is more clinically relevant for graft outcome than neat IgG MFI. IgG titers, subclass, and other complement assays have now been studied to determine if they are more relevant. Only IgG3 and possibly C3d fixation have shown added value to C1q for outcome correlation. Direct parallel titer comparisons of C1q and IgG are lacking and the correlation is unknown. SUMMARY: Overall, results confirm the correlation with C1q+ donor-specific antibody (DSA) for AMR and graft loss. The association is stronger posttransplant. C1q+ de novo antibody appears to be especially detrimental portending graft loss in about 1-2.5 years post detection. Recommendations to biopsy and treat at time of de novo C1q+ antibody detection have been suggested by several groups.


Assuntos
Complemento C1q/imunologia , Rejeição de Enxerto/imunologia , Antígenos HLA/imunologia , Isoanticorpos/imunologia , Humanos
9.
J Burn Care Res ; 38(3): 169-173, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27801681

RESUMO

Skin allografts are the gold standard in temporary burn wound coverage, but allografts are hypothesized to place a high antigenic load on recipients. This project aims to determine the degree of human leukocyte antigen sensitization in burn patients treated with allografts. Serum was obtained from nine adult, nontransfused, and nontransplanted burn patients treated with allografts. Group 1 included patients tested in the acute burn period, while group 2 included different patients tested months to years after injury. A calculated panel reactive antibody (cPRA) percent was assessed for each patient, and data for a control group of 92 adult nontransplanted males were used for comparison. Each patient received allografts from an average 3.55 ± 1.24 different donors. cPRA in group 1 was lower than in group 2 (6 ± 12% vs 42 ± 33%, P = .08). cPRA in the study group was significantly higher than in the control group (26 ± 31% vs 8 ± 17%, P = .0075). Burn patients who receive skin allograft demonstrate increased immunological sensitization compared with unsensitized controls. Detection of human leukocyte antigen antibody is lower in the acute burn period than months to years after injury. Increased sensitization may ultimately limit burn patients' candidacy for vascularized composite allotransplantation or decrease success of these procedures.


Assuntos
Queimaduras/imunologia , Queimaduras/terapia , Transplante de Pele , Alotransplante de Tecidos Compostos Vascularizados , Adulto , Idoso , Aloenxertos , Feminino , Rejeição de Enxerto/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento
10.
Pediatr Transplant ; 18(8): E280-2, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25174602

RESUMO

The proteasome inhibitor bortezomib has been used with variable success in the treatment of AMR following heart transplant. There is limited experience with this agent as a pretransplant desensitizing therapy. We report a case of successful HLA desensitization with a bortezomib-based protocol prior to successful heart transplantation. A nine-yr-old boy with dilated cardiomyopathy, not initially sensitized to HLA (cPRA of zero), required three days of ECMO, followed by implantation of a Heartmate II LVAD. Within six wk, the patient developed de novo class I IgG and C1q complement-fixing HLA antibodies with a cPRA of 100%. Two doses of IVIG (2 g/kg) failed to reduce antibody levels, although two courses of a novel desensitization protocol consisting of rituximab (375 mg/m(2) ), bortezomib (1.3 mg/m(2)  × 5 doses), and plasmapheresis reduced his cPRA to 0% and 87% by the C1q and IgG assays, respectively. He underwent heart transplantation nearly two months later. The patient is now >one yr post-transplant, is free of both AMR and ACR, and has no detectable donor-specific antibodies by IgG or C1q. Proteasome inhibition with bortezomib and plasmapheresis may be an effective therapy for HLA desensitization pretransplant.


Assuntos
Ácidos Borônicos/uso terapêutico , Cardiomiopatia Dilatada/cirurgia , Rejeição de Enxerto/prevenção & controle , Antígenos HLA/imunologia , Transplante de Coração , Inibidores de Proteassoma/uso terapêutico , Pirazinas/uso terapêutico , Condicionamento Pré-Transplante/métodos , Anticorpos Monoclonais Murinos/uso terapêutico , Bortezomib , Criança , Terapia Combinada , Quimioterapia Combinada , Rejeição de Enxerto/imunologia , Humanos , Fatores Imunológicos/uso terapêutico , Isoanticorpos/imunologia , Masculino , Plasmaferese , Rituximab
11.
Transpl Int ; 27(12): 1235-43, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24990476

RESUMO

Terminal complement blockade has been shown to decrease the incidence of early acute antibody-mediated rejection (eAMR) in the first month after positive cross-match kidney transplant recipients, yet some patients still develop eAMR. The current study investigated possible mechanisms of eAMR despite eculizumab treatment. Of the 26 patients treated with eculizumab, two developed clinical eAMR and another patient developed histologic signs of eAMR without graft dysfunction ('subclinical eAMR'). Twenty-three did not have histologic injury on early surveillance biopsies. All 26 patients had therapeutic levels of eculizumab and showed complete blockade of complement in hemolytic assays. High levels of donor-specific alloantibody (DSA) including total IgG, IgG3, and C1q+ DSA were present in patients with and without eAMR, and none correlated well with eAMR. In contrast, IgM DSA was present in only four patients after transplantation: the two patients with clinical eAMR, one patient with subclinical AMR, and one patient without eAMR (P = 0.006 correlation with eAMR). Both clinical eAMR episodes were easily treated with plasma exchange which removed IgM more completely and rapidly than IgG, resulting in normalization of function and histology. These data suggest a possible role of antidonor IgM DSA in the pathogenesis of eAMR in patients treated with terminal complement blockade (ClinicalTrials.gov Identifier: NCT00670774).


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Complemento C5/antagonistas & inibidores , Rejeição de Enxerto/imunologia , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Isoanticorpos/imunologia , Transplante de Rim , Anticorpos Monoclonais Humanizados/farmacologia , Complemento C1q/imunologia , Complemento C5/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Rejeição de Enxerto/terapia , Antígenos HLA/imunologia , Teste de Histocompatibilidade , Humanos , Troca Plasmática , Falha de Tratamento
12.
Nephrol Dial Transplant ; 28(11): 2908-18, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24009295

RESUMO

BACKGROUND: Sensitization to human leukocyte antigen (HLA) from red blood cell (RBC) transfusion is poorly quantified and is based on outdated, insensitive methods. The objective was to evaluate the effect of transfusion on the breadth, magnitude and specificity of HLA antibody formation using sensitive and specific methods. METHODS: Transfusion, demographic and clinical data from the US Renal Data System were obtained for patients on dialysis awaiting primary kidney transplant who had ≥ 2 HLA antibody measurements using the Luminex single-antigen bead assay. One cohort included patients with a transfusion (n = 50) between two antibody measurements matched with up to four nontransfused patients (n = 155) by age, sex, race and vintage (time on dialysis). A second crossover cohort (n = 25) included patients with multiple antibody measurements before and after transfusion. We studied changes in HLA antibody mean fluorescence intensity (MFI) and calculated panel reactive antibody (cPRA). RESULTS: In the matched cohort, 10 of 50 (20%) transfused versus 6 of 155 (4%) nontransfused patients had a ≥ 10 HLA antibodies increase of >3000 MFI (P = 0.0006); 6 of 50 (12%) transfused patients had a ≥ 30 antibodies increase (P = 0.0007). In the crossover cohort, the number of HLA antibodies increasing >1000 and >3000 MFI was higher in the transfused versus the control period, P = 0.03 and P = 0.008, respectively. Using a ≥ 3000 MFI threshold, cPRA significantly increased in both matched (P = 0.01) and crossover (P = 0.002) transfused patients. CONCLUSIONS: Among prospective primary kidney transplant recipients, RBC transfusion results in clinically significant increases in HLA antibody strength and breadth, which adversely affect the opportunity for future transplant.


Assuntos
Anticorpos/sangue , Formação de Anticorpos/imunologia , Transfusão de Sangue , Antígenos HLA/imunologia , Falência Renal Crônica/imunologia , Transplante de Rim , Anticorpos/imunologia , Estudos Cross-Over , Feminino , Humanos , Falência Renal Crônica/sangue , Falência Renal Crônica/cirurgia , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Diálise Renal , Listas de Espera
13.
Methods Mol Biol ; 1034: 305-11, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23775744

RESUMO

Solid phase Luminex(®) and flow cytometric single antigen bead assays offer exquisite sensitivity and specificity for HLA antibody detection. Unlike the historical complement-dependent cytotoxicity (CDC) method, these assays do not distinguish complement fixing from non-complement fixing antibody, the former of which are considered the most clinically relevant in the peri-transplant period. This chapter describes a novel solid phase C1q binding assay to distinguish HLA antibodies that can bind the first component of complement (C1q). These antibodies have the capacity to initiate the complement cascade irrespective of whether that actually occurs. The C1q assay detects many more complement fixing antibodies than are observed by the less sensitive and less specific CDC assay.


Assuntos
Anticorpos/imunologia , Complemento C1q/imunologia , Antígenos HLA/imunologia , Transplante Homólogo , Especificidade de Anticorpos , Testes de Fixação de Complemento , Citometria de Fluxo , Rejeição de Enxerto/imunologia , Humanos , Biologia Molecular/métodos , Transplante
14.
Transplantation ; 95(1): 19-47, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23238534

RESUMO

BACKGROUND: The introduction of solid-phase immunoassay (SPI) technology for the detection and characterization of human leukocyte antigen (HLA) antibodies in transplantation while providing greater sensitivity than was obtainable by complement-dependent lymphocytotoxicity (CDC) assays has resulted in a new paradigm with respect to the interpretation of donor-specific antibodies (DSA). Although the SPI assay performed on the Luminex instrument (hereafter referred to as the Luminex assay), in particular, has permitted the detection of antibodies not detectable by CDC, the clinical significance of these antibodies is incompletely understood. Nevertheless, the detection of these antibodies has led to changes in the clinical management of sensitized patients. In addition, SPI testing raises technical issues that require resolution and careful consideration when interpreting antibody results. METHODS: With this background, The Transplantation Society convened a group of laboratory and clinical experts in the field of transplantation to prepare a consensus report and make recommendations on the use of this new technology based on both published evidence and expert opinion. Three working groups were formed to address (a) the technical issues with respect to the use of this technology, (b) the interpretation of pretransplantation antibody testing in the context of various clinical settings and organ transplant types (kidney, heart, lung, liver, pancreas, intestinal, and islet cells), and (c) the application of antibody testing in the posttransplantation setting. The three groups were established in November 2011 and convened for a "Consensus Conference on Antibodies in Transplantation" in Rome, Italy, in May 2012. The deliberations of the three groups meeting independently and then together are the bases for this report. RESULTS: A comprehensive list of recommendations was prepared by each group. A summary of the key recommendations follows. Technical Group: (a) SPI must be used for the detection of pretransplantation HLA antibodies in solid organ transplant recipients and, in particular, the use of the single-antigen bead assay to detect antibodies to HLA loci, such as Cw, DQA, DPA, and DPB, which are not readily detected by other methods. (b) The use of SPI for antibody detection should be supplemented with cell-based assays to examine the correlations between the two types of assays and to establish the likelihood of a positive crossmatch (XM). (c) There must be an awareness of the technical factors that can influence the results and their clinical interpretation when using the Luminex bead technology, such as variation in antigen density and the presence of denatured antigen on the beads. Pretransplantation Group: (a) Risk categories should be established based on the antibody and the XM results obtained. (b) DSA detected by CDC and a positive XM should be avoided due to their strong association with antibody-mediated rejection and graft loss. (c) A renal transplantation can be performed in the absence of a prospective XM if single-antigen bead screening for antibodies to all class I and II HLA loci is negative. This decision, however, needs to be taken in agreement with local clinical programs and the relevant regulatory bodies. (d) The presence of DSA HLA antibodies should be avoided in heart and lung transplantation and considered a risk factor for liver, intestinal, and islet cell transplantation. Posttransplantation Group: (a) High-risk patients (i.e., desensitized or DSA positive/XM negative) should be monitored by measurement of DSA and protocol biopsies in the first 3 months after transplantation. (b) Intermediate-risk patients (history of DSA but currently negative) should be monitored for DSA within the first month. If DSA is present, a biopsy should be performed. (c) Low-risk patients (nonsensitized first transplantation) should be screened for DSA at least once 3 to 12 months after transplantation. If DSA is detected, a biopsy should be performed. In all three categories, the recommendations for subsequent treatment are based on the biopsy results. CONCLUSIONS: A comprehensive list of recommendations is provided covering the technical and pretransplantation and posttransplantation monitoring of HLA antibodies in solid organ transplantation. The recommendations are intended to provide state-of-the-art guidance in the use and clinical application of recently developed methods for HLA antibody detection when used in conjunction with traditional methods.


Assuntos
Antígenos HLA/imunologia , Isoanticorpos/sangue , Transplante de Órgãos , Complemento C1q/análise , Complemento C4b , Proteínas do Sistema Complemento/imunologia , Citotoxicidade Imunológica , Citometria de Fluxo/métodos , Humanos , Imunoensaio , Isoanticorpos/imunologia , Fragmentos de Peptídeos/sangue , Guias de Prática Clínica como Assunto
15.
Curr Opin Organ Transplant ; 17(4): 409-15, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22710388

RESUMO

PURPOSE OF REVIEW: Complement-fixing human leukocyte antigen (HLA) antibodies are a contraindication to solid organ transplant. Newer solid phase assays for HLA antibody definition have created treatment quandaries because many more antibodies are detected by these methods. It is unclear which of the antibodies identified are clinically relevant as all IgG-binding antibodies are detected whether they can fix complement or not. RECENT FINDINGS: Two methods have been developed to assess complement-fixing capability in the solid phase assays: C4d and C1q. These assays, especially the sensitive C1q method, have been reported to more closely correlate with renal and cardiac graft dysfunction, rejection, and graft failure than antibodies detected only by the traditional IgG method. Additionally, the C1q method can be used to predict and monitor desensitization status pre and posttransplant in patients being treated with intravenous immunoglobulin. SUMMARY: The availability of these complement-fixing assays provides new tools for making treatment decisions by discriminating antibodies with known clinical relevance and to assess the clinical relevance of binding antibodies that cannot fix complement. The assays also provide a means of assessing when to transplant a patient undergoing desensitization or when to discontinue augmented immunosuppression for resolving antibody-mediated rejection.


Assuntos
Anticorpos/imunologia , Proteínas do Sistema Complemento/análise , Rejeição de Enxerto/diagnóstico , Antígenos HLA/imunologia , Anticorpos/sangue , Complemento C1q/análise , Complemento C1q/imunologia , Complemento C4b/imunologia , Proteínas do Sistema Complemento/imunologia , Rejeição de Enxerto/imunologia , Humanos , Fragmentos de Peptídeos/sangue , Fragmentos de Peptídeos/imunologia
16.
Pediatr Transplant ; 16(1): 12-7, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22093755

RESUMO

Long-term outcomes following renal transplantation remain disappointing. Recently, interest has focused on the antibody-mediated component of allograft injury and the deleterious effects of DSA. We applied a novel C1q solid-phase assay in parallel with the standard IgG SAB assay to identify DSA with the potential to activate complement by binding C1q. Among 193 consecutive renal transplants at our center, 19.2% developed de novo DSA following transplantation. Of the patients with DSA, 43% had antibodies that bound C1q in vitro [C1q+ DSA]. Patients with C1q+ DSA were more likely to develop allograft loss than patients with DSA that did not bind C1q (46.7% vs. 15%; p = 0.04); patients with C1q+ DSA were nearly six times more likely to lose their transplant than those with C1q- DSA. Additionally, patients with C1q+ DSA who underwent allograft biopsy were more likely to demonstrate C4d deposition (50% vs. 8%; p = 0.03) and meet criteria for acute rejection (60% vs. 17%; p = 0.02) when compared with patients with DSA that did not bind C1q. These data suggest that DSA with the ability to activate complement, as determined by this novel C1q assay, are associated with greater risk of acute rejection and allograft loss.


Assuntos
Complemento C1q/química , Rejeição de Enxerto , Transplante de Rim/imunologia , Transplante Homólogo/imunologia , Adolescente , Biópsia , Criança , Pré-Escolar , Ativação do Complemento , Testes de Fixação de Complemento , Feminino , Humanos , Imunoglobulina G/química , Masculino , Insuficiência Renal/terapia , Estudos Retrospectivos , Resultado do Tratamento
17.
Transfusion ; 51(12): 2611-8, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21615749

RESUMO

BACKGROUND: Immune refractoriness to platelet (PLT) transfusion is primarily due to HLA antibody. Patients at our institution are identified as refractory due to HLA by a Luminex-based immunoglobulin (Ig)G-single-antigen-bead (SAB) assay, but in highly sensitized patients, antigen-negative compatible donors cannot be found due to the high sensitivity of the IgG-SAB method. We developed an assay that detects only HLA antibodies binding the first complement component (C1q). We hypothesized that the C1q-SAB method might be more relevant than the IgG-SAB method because the antibodies identified may activate the complement cascade causing PLT destruction. STUDY DESIGN AND METHODS: Thirteen highly sensitized refractory patients received 177 PLT units incompatible by the IgG-SAB method. They were retrospectively retested by the C1q-SAB method. Calculated percent reactive antibody (CPRA) and HLA antibody specificities were compared between the two methods and corrected count increment (CCI) values were analyzed. Additionally the impact of ABO compatibility on CCI responses was evaluated. RESULTS: The mean CPRA value was significantly lower by C1q-SAB (60%) than by IgG-SAB (94%; p < 0.05). Patients showed significantly better CCI (10.6 × 10(9) ± 0.8 × 10(9) /L) with C1q-compatible (n = 134) than with C1q-incompatible PLTs (n = 43) (2.5 × 10(9) ± 0.9 × 10(9) /L/m(2) ; p < 0.0001). ABO compatibility did not significantly impact the CCI values (p < 0.0001). Our results show that 75% of PLT units previously considered incompatible were actually compatible. CONCLUSION: For highly refractory patients to PLT transfusion, the C1q-based SAB binding assay may be a better method for identifying clinically relevant HLA antibodies and selecting PLT units that will result in acceptable CCI.


Assuntos
Bioensaio/métodos , Plaquetas , Complemento C1q/química , Antígenos HLA/imunologia , Imunoglobulina G , Isoanticorpos , Sistema ABO de Grupos Sanguíneos/sangue , Sistema ABO de Grupos Sanguíneos/imunologia , Adulto , Idoso , Seleção do Doador/métodos , Feminino , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Isoanticorpos/sangue , Isoanticorpos/imunologia , Masculino , Pessoa de Meia-Idade
18.
Transplantation ; 91(3): 342-7, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21116220

RESUMO

BACKGROUND: Human leukocyte antigen (HLA) antibodies, especially those that fix complement, are associated with antibody-mediated rejection and graft failure. The C1q assay on single antigen beads detects a subset of HLA antibodies that can fix complement and precede C4d deposition. The aim of this study was to determine whether C1q-fixing antibodies distinguish de novo donor-specific antibodies (DSA) that are clinically relevant and harmful. METHODS: We retrospectively studied 31 of 274 kidney transplant recipients who had pretransplant and concurrent biopsy and serum specimens, 13 with C4d-positive and 18 with C4d-negative staining. We measured IgG and C1q DSA pretransplant and at the time of biopsy using single antigen bead assays. We identified 13 recipients who developed de novo DSA by IgG or C1q and examined associations with C4d deposition, transplant glomerulopathy, and graft failure. RESULTS: Testing for DSA by IgG is more sensitive for C4d deposition (IgG: 100%, 95% confidence interval [CI] 0.60-1; C1q: 75%, 95% CI 0.36-0.96). Testing for DSA by C1q is more specific for transplant glomerulopathy (C1q: 81%, 95% CI 0.57-0.94; IgG: 67%, 95% CI 0.43-0.85) and graft loss (C1q: 79%, 95% CI 0.54-0.93; IgG: 63%, 95% CI 0.39-0.83). Absence of de novo DSA by IgG and C1q has a high negative predictive value for the absence of C4d deposition (IgG: 100%, 95% CI 0.73-1; C1q: 88%, 95% CI 0.62-0.98), transplant glomerulopathy (IgG: 100%, 95% CI 0.73-1; C1q: 100%, 95% CI 0.77-1), and graft failure (IgG: 86%, 95% CI 0.56-0.97; C1q: 88%, 95% CI 0.62-0.98). CONCLUSION: Monitoring patients with the C1q assay, which detects antibodies that fix complement, offers a minimally invasive means of identifying patients at risk for transplant glomerulopathy and graft loss.


Assuntos
Anticorpos/sangue , Complemento C1q/imunologia , Rejeição de Enxerto/epidemiologia , Antígenos HLA/imunologia , Nefropatias/epidemiologia , Transplante de Rim , Adulto , Biomarcadores/sangue , Biópsia , Complemento C4b/metabolismo , Feminino , Seguimentos , Rejeição de Enxerto/sangue , Humanos , Estimativa de Kaplan-Meier , Rim/metabolismo , Rim/patologia , Nefropatias/sangue , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/metabolismo , Projetos Piloto , Valor Preditivo dos Testes , Estudos Retrospectivos , Fatores de Risco , Sensibilidade e Especificidade
20.
Transplantation ; 78(2): 181-5, 2004 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-15280674

RESUMO

During the past few decades, much of the experimental and clinical effort in solid-organ transplantation has been directed toward ameliorating or abrogating T-cell-mediated responses. As a result, universally understood and accepted nomenclature and diagnostic criteria have evolved. Humoral immunity in transplantation has yet to undergo a similar renaissance. Readers of transplant journals regularly find it difficult and often impossible to interpret data on the diagnosis and management of antibody-mediated rejection. The Antibody Working Group was assembled in an attempt to provide guidelines for the standardization of nomenclature, diagnostic criteria, reporting, antibody profiling, and risk assessment.


Assuntos
Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Terapia de Imunossupressão/métodos , Isoanticorpos/imunologia , Linfócitos T/imunologia , Humanos , Modelos Imunológicos , Condicionamento Pré-Transplante/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA