Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Robert A. Bray is active.

Publication


Featured researches published by Robert A. Bray.


Anatomical Record-advances in Integrative Anatomy and Evolutionary Biology | 2001

Human reserve pluripotent mesenchymal stem cells are present in the connective tissues of skeletal muscle and dermis derived from fetal, adult, and geriatric donors

Henry E. Young; Timothy A. Steele; Robert A. Bray; John Hudson; Julie A. Floyd; Kristina C. Hawkins; Karen Thomas; Chris Edwards; Jeremy Cuzzourt; Mary Duenzl; Paul A. Lucas; Asa C. Black

This study details the profile of 13 cell surface cluster differentiation markers on human reserve stem cells derived from connective tissues. Stem cells were isolated from the connective tissues of dermis and skeletal muscle derived from fetal, mature, and geriatric humans. An insulin/dexamethasone phenotypic bioassay was used to determine the identity of the stem cells from each population. All populations contained lineage‐committed myogenic, adipogenic, chondrogenic, and osteogenic progenitor stem cells as well as lineage‐uncommitted pluripotent stem cells capable of forming muscle, adipocytes, cartilage, bone, fibroblasts, and endothelial cells. Flow cytometric analysis of adult stem cell populations revealed positive staining for CD34 and CD90 and negative staining for CD3, CD4, CD8, CD11c, CD33, CD36, CD38, CD45, CD117, Glycophorin‐A, and HLA DR‐II. Anat Rec 264:51–62, 2001.


Transplantation | 2013

Consensus guidelines on the testing and clinical management issues associated with HLA and non-HLA antibodies in transplantation.

Brian D. Tait; Caner Süsal; Howard M. Gebel; Peter Nickerson; Andrea A. Zachary; Frans H.J. Claas; Elaine F. Reed; Robert A. Bray; Patricia Campbell; Jeremy R. Chapman; P. Toby Coates; Robert B. Colvin; Emanuele Cozzi; Ilias I.N. Doxiadis; S. V. Fuggle; John S. Gill; Nils Lachmann; Thalachallour Mohanakumar; Nicole Suciu-Foca; Suchitra Sumitran-Holgersson; Kazunari Tanabe; Craig J. Taylor; Dolly B. Tyan; Angela C Webster; Adriana Zeevi; Gerhard Opelz

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.


American Journal of Transplantation | 2003

Pre-Transplant Assessment of Donor-Reactive, HLA-Specific Antibodies in Renal Transplantation: Contraindication vs. Risk

Howard M. Gebel; Robert A. Bray; Peter Nickerson

In renal transplantation, a positive cytotoxic crossmatch between donor cells and recipient serum is associated with early rejection or graft loss and was the driving force behind the establishment of HLA laboratories. Initially, crossmatches were performed by relatively insensitive techniques [e.g. leukoagglutination and direct complement-dependent cytotoxicity (CDC) of target cells]. A negative result justified proceeding, while a positive crossmatch was considered a contraindication to renal transplantation. However, the underlying premises driving this practice, namely that (i) all positive reactions were the result of relevant (i.e. HLA) antibodies that could lead to allograft rejection; and (ii) all negative reactions predicted long-term graft survival were known to be incorrect. From its first clinical description, the simple complementdependent assay was recognized as neither sufficiently specific nor sensitive to identify all relevant antibodies. Over time, more sensitive and specific lymphocyte crossmatch assays were developed that effectively decreased the incidence of early antibody-mediated rejection.


Transplantation | 2000

Sensitization and sensitivity: defining the unsensitized patient.

Howard M. Gebel; Robert A. Bray

BACKGROUND Since the landmark studies of Patel and Terasaki in the late 1960s, pretransplant cross-matching has been performed by HLA laboratories on a 24-hr/7-day basis. In fact, regulating agencies such as the American Society for Histocompatibility and Immunogenetics and the United Network for Organ Sharing have mandated prospective crossmatching for selected solid organ transplants. However, two recent publications (Transplantation 1998; 66: 1833; and Transplantation 1998; 66: 1835) have suggested a change to this approach. Specifically, those authors advocate the transplantation of non-sensitized individuals without a final prospective cross-match as a means to reduce cold ischemia time and the incidence of delayed graft function. Such considerations were predicated upon results generated by cytotoxicity-based antibody screening. We and others, however, have reported that a flow cytometric-based assay is a more sensitive method to detect alloantibodies than cytotoxicity. Furthermore, an increasing number of reports document that graft survival is improved among patients whose final flow cytometric crossmatches were negative compared to patients with positive flow cytometric crossmatches. Although we agree that it is reasonable to transplant truly non-sensitized patients without a prospective final crossmatch, our data demonstrate that a large number of patients deemed non-sensitized by cytotoxicity-based antibody assessment are, in fact, sensitized. METHODS Panel-reactive antibody (PRA) testing was performed with 703 sera from 527 patients. The patient population consisted of individuals awaiting either renal or cardiac transplantation. PRA evaluations were performed using lymphocyte cytotoxicity (antiglobulin-enhanced, complement-dependent cytotoxicity [AHG-CDC]) or assays (enzyme-linked immunosorbent assay [ELISA]; flow cytometry) in which solubilized HLA molecules were affixed to solid phase matrices. RESULTS PRA activity in 264 sera from 88 patients was evaluated by AHG-CDC, ELISA, and flow cytometry. Results among the three methods were concordant for 83% of these sera. Discordant results occurred with 32 samples and demonstrated a distinct hierarchy in the sensitivity of the three techniques to detect alloantibodies. None of the 32 sera were positive by AHG-CDC, 20/32 were positive by ELISA, and 32/32 were positive by flow cytometry. Subsequent studies revealed that, among 527 patients, 302 (57%) exhibited 0% PRA by AHG-CDC. Of these 302 AHG-CDC-negative patients, 76 (25%) had class I or class II antibodies detectable using a flow cytometric approach. Within the AHG-CDC-negative/flow cytometric-positive patients, PRA values exhibited a wide range (6-99%) for both class I and class II antibodies. The average PRA was 27% and 38% for class I and II, respectively. Retrospective flow cytometric crossmatches performed for 30 recipients of cardiac allografts whose AHG-CDC PRA were 0% revealed that 11/30 crossmatches were positive. CONCLUSIONS The concept of transplanting non-sensitized patients without a prospective final crossmatch is appealing and, if bona fide, clearly makes sense. However, our data demonstrate that how a patient is deemed non-sensitized is critical. The difference between AHG- and flow cytometric-based PRA testing is significant and can result in transplantation of alloimmunized patients considered to be non-sensitized. Therefore, we recommend that, if a transplant center chooses to forego a prospective final crossmatch, the decision to do so should be based on methods more sensitive than AHG-CDC.


American Journal of Transplantation | 2006

Transplanting the highly sensitized patient: The emory algorithm.

Robert A. Bray; J. D. L. Nolen; Christian P. Larsen; Thomas C. Pearson; Kenneth A. Newell; K. Kokko; Antonio Guasch; P. Tso; J. B. Mendel; Howard M. Gebel

Renal transplant patients sensitized to HLA antigens comprise nearly one‐third of the UNOS wait‐list and receive 14% of deceased donor (DD) transplants, a rate half that of unsensitized patients. Between 1999 and 2003, we performed 492 adult renal transplants from DD; 120 patients (∼25%) had a panel reactive antibody (PRA) of >30%, with nearly half (n = 58) having a PRA of >80%. Our approach is based upon high‐resolution solid‐phase HLA antibody analysis to identify class I/II antibodies and a ‘virtual crossmatch’ to predict compatible donor/recipient combinations. Recipients are excluded from the United Network for Organ Sharing match run if donors possess unacceptable antigens. Thus, when sensitized patients appear on the match run, they have a high probability of a negative final crossmatch. Here, we describe our 5‐year experience with this approach. Five‐year graft survival ranged from 66% to 70% among unsensitized (n = 272), moderately sensitized (PRA < 30%, n = 100) and highly sensitized (>30% PRA; n = 120) patients, equal to the average national graft survival (65.7%). The application of this approach (the Emory Algorithm) provides a logical and systematic approach to improve the access of sensitized patients to DD organs and promote more equitable allocation to a highly disadvantaged group of patients awaiting renal transplantation.


Blood | 2010

The detection of donor-directed, HLA-specific alloantibodies in recipients of unrelated hematopoietic cell transplantation is predictive of graft failure

Stephen Spellman; Robert A. Bray; Sandra Rosen-Bronson; Michael Haagenson; John P. Klein; Susan Flesch; Cynthia Vierra-Green; Claudio Anasetti

Donor-directed human leukocyte antigen (HLA)-specific allo-antibodies (DSAs) cause graft failure in animal models of hematopoietic stem cell transplantation (HCT). Archived pretransplantation sera from graft failure patients (n = 37) and a matched case-control cohort (n = 78) were tested to evaluate the role of DSAs in unrelated donor HCT. Controls were matched for disease, disease status, graft type, patient age, and transplantation year. Patients had acute myeloid leukemia, acute lymphoblastic leukemia, chronic myelogenous leukemia, or myelodysplastic syndrome; 98% received myeloablative conditioning regimens 100% received T-replete grafts, 97% received marrow, 95% HLA-mismatched, and 97% received calcineurin-based graft-versus-host disease prophylaxis. Among the 37 failed transplantations, 9 (24%) recipients possessed DSAs against HLA-A, B, and/or DP, compared with only 1 (1%) of 78 controls. Therefore, the presence of DSAs was significantly associated with graft failure (odds ratio = 22.84; 95% confidence interval, 3.57-infinity; P < .001). These results indicate that the presence of pretransplantation DSAs in recipients of unrelated donor HCT is associated with failed engraftment and should be considered in HCT donor selection.


Transplantation | 2000

Flow cytometric detection of HLA-specific antibodies as a predictor of heart allograft rejection

Anat R. Tambur; Robert A. Bray; Steven K. Takemoto; Mary Mancini; Maria Rosa Costanzo; J. Kobashigawa; Corby L. D'amico; Kirk R. Kanter; Alexandria M. Berg; J. David Vega; Andrew L. Smith; Anthony L. Roggero; John W. Ortegel; Lisa Wilmoth-Hosey; J. Michael Cecka; Howard M. Gebel

BACKGROUND Historically, panel reactive antibody (PRA) analysis to detect HLA antibodies has been performed using cell-based complement-dependent cytotoxicity (CDC) techniques. Recently, a flow cytometric procedure (FlowPRA) was introduced as an alternative approach to detect HLA antibodies. The flow methodology, using a solid phase matrix to which soluble HLA class I or class II antigens are attached is significantly more sensitive than CDC assays. However, the clinical relevance of antibodies detected exclusively by FlowPRAhas not been established. In this study of cardiac allograft recipients, FlowPRA was performed on pretransplant sera with no detectable PRA activity as assessed by CDC assays. FlowPRA antibody activity was then correlated with clinical outcome. METHODS PRA analysis by anti-human globulin enhanced (AHG) CDC and FlowPRA was performed on sera corresponding to final cross-match specimens from 219 cardiac allograft recipients. In addition, sera collected 3-6 months posttransplant from 91 patients were evaluated. The presence or absence of antibodies was correlated with episodes of rejection and patient survival. A rejection episode was considered to have occurred based on treatment with antirejection medication and/or histology. RESULTS By CDC, 12 patients (5.5%) had pretransplant PRA >10%. In contrast, 72 patients (32.9%) had pretransplant anti-HLA antibodies detectable by FlowPRA (34 patients with only class I antibodies; 7 patients with only class II antibodies; 31 patients with both class I and class II antibodies). A highly significant association (P<0.001) was observed between pretransplant HLA antibodies detected by FlowPRA and episodes of rejection that occurred during the first posttransplant year. Fifteen patients died within the first year posttransplant. Of nine retrospective flow cytometric cross-matches that were performed, two were in recipients who had no pretransplant antibodies detectable by FlowPRA. Both of these cross-matches were negative. In contrast, five of seven cross-matches were positive among recipients who had FlowPRA detectable pretransplant antibodies. Posttransplant serum specimens from 91 patients were also assessed for antibodies by FlowPRA. Among this group, 58 patients had FlowPRA antibodies and there was a trend (although not statistically significant) for a biopsy documented episode of rejection to have occurred among patients with these antibodies. CONCLUSIONS Collectively, our data suggest that pre- and posttransplant HLA antibodies detectable by FlowPRA and not AHG-CDC identify cardiac allograft recipients at risk for rejection. Furthermore, a positive donor reactive flow cytometric cross-match is significantly associated with graft loss. Thus, we believe that detection and identification of HLA-specific antibodies can be used to stratify patients into high and low risk categories. An important observation of this study is that in the majority of donor:recipient pairs, pretransplant HLA antibodies were not directed against donor antigens. We speculate that these non-donor-directed antibodies are surrogate markers that correspond to previous T cell activation. Thus, the rejection episodes that occur in these patients are in response to donor-derived MHC peptides that share cryptic determinants with the HLA antigens that initially sensitized the patient.


American Journal of Transplantation | 2011

Revisiting Traditional Risk Factors for Rejection and Graft Loss after Kidney Transplantation

Ty B. Dunn; Harriet Noreen; K. J. Gillingham; David Maurer; O. G. Ozturk; Timothy L. Pruett; Robert A. Bray; Howard M. Gebel; Arthur J. Matas

Single‐antigen bead (SAB) testing permits reassessment of immunologic risk for kidney transplantation. Traditionally, high panel reactive antibody (PRA), retransplant and deceased donor (DD) grafts have been associated with increased risk. We hypothesized that this risk was likely mediated by (unrecognized) donor‐specific antibody (DSA). We grouped 587 kidney transplants using clinical history and single‐antigen bead (SAB) testing of day of transplant serum as (1) unsensitized; PRA = 0 (n = 178), (2) third‐party sensitized; no DSA (n = 363) or (3) donor sensitized; with DSA (n = 46), and studied rejection rates, death‐censored graft survival (DCGS) and risk factors for rejection. Antibody‐mediated rejection (AMR) rates were increased with DSA (p < 0.0001), but not with panel reactive antibody (PRA) in the absence of DSA. Cell‐mediated rejection (CMR) rates were increased with DSA (p < 0.005); with a trend to increased rates when PRA>0 in the absence of DSA (p = 0.08). Multivariate analyses showed risk factors for AMR were DSA, worse HLA matching, and female gender; for CMR: DSA, PRA>0 and worse HLA matching. AMR and CMR were associated with decreased DCGS. The presence of DSA is an important predictor of rejection risk, in contrast to traditional risk factors. Further development of immunosuppressive protocols will be facilitated by stratification of rejection risk by donor sensitization.


Transplantation | 2004

Persistence of low levels of alloantibody after desensitization in crossmatch-positive living-donor kidney transplantation.

James M. Gloor; Steven R. DeGoey; Nancy A. Ploeger; Howard Gebel; Robert A. Bray; S. Breanndan Moore; Patrick G. Dean; Mark D. Stegall

Background: Desensitization protocols have been developed to allow successful kidney transplantation in sensitized recipients. However, a detailed analysis of the impact of these protocols on alloantibody has not been performed. Methods: We studied 12 living-donor kidney-transplant recipients with positive antihuman globulin-enhanced complement dependent cytotoxicity (AHG-CDC) crossmatches against their donors. Using a variety of crossmatch techniques and single-antigen flowbeads (SAFBs), we characterized the specificity and amount of alloantibody at baseline before desensitization, after desensitization (using plasmapheresis followed by 100 mg/kg intravenous immunoglobulin, and anti-CD20 antibody), and 4 months after transplantation (after splenectomy and on maintenance immunosuppression). Results: All 12 patients with a positive baseline AHG-CDC crossmatch were AHG-CDC crossmatch negative at the time of transplant (after desensitization). However, despite desensitization, the majority of patients had low-level donor-specific alloantibodies demonstrable on the day of transplantation by both flow crossmatch (FXM 8/12) and SAFBs (10/11). Four months after transplantation, no patient had a positive AHG-CDC crossmatch, but again the majority had persistent low levels of donor-specific alloantibodies by FXM (6/12) and SAFBs (9/11). No patient experienced hyperacute rejection, and the persistence of low levels of donor-specific alloantibodies did not correlate with the development of humoral rejection in the early posttransplant period. Conclusions: Despite desensitization, a majority of positive crossmatch transplant recipients demonstrate low levels of donor-specific alloantibodies both on the day of transplant and 4 months after transplantation. The impact of these antibodies appears to be minimal early after transplant, but their long-term significance bears further study.


American Journal of Transplantation | 2013

Comprehensive assessment and standardization of solid phase multiplex-bead arrays for the detection of antibodies to HLA.

Elaine F. Reed; Ping Rao; Zilu Zhang; Howard M. Gebel; Robert A. Bray; Indira Guleria; John G. Lunz; Thalachallour Mohanakumar; Peter Nickerson; Anat R. Tambur; Adriana Zeevi; Peter S. Heeger; David W. Gjertson

Solid phase multiplex‐bead arrays for the detection and characterization of HLA antibodies provide increased sensitivity and specificity compared to conventional lymphocyte‐based assays. Assay variability due to inconsistencies in commercial kits and differences in standard operating procedures (SOP) hamper comparison of results between laboratories. The Clinical Trials in Organ Transplantation Antibody Core Laboratories investigated sources of assay variation and determined if reproducibility improved through utilization of SOP, common reagents and normalization algorithms. Ten commercial kits from two manufacturers were assessed in each of seven laboratories using 20 HLA reference sera. Implementation of a standardized (vs. a nonstandardized) operating procedure greatly reduced MFI variation from 62% to 25%. Although laboratory agreements exceeded 90% (R2), small systematic differences were observed suggesting center specific factors still contribute to variation. MFI varied according to manufacturer, kit, bead type and lot. ROC analyses showed excellent consistency in antibody assignments between manufacturers (AUC > 0.9) and suggested optimal cutoffs from 1000 to 1500 MFI. Global normalization further reduced MFI variation to levels near 20%. Standardization and normalization of solid phase HLA antibody tests will enable comparison of data across laboratories for clinical trials and diagnostic testing.

Collaboration


Dive into the Robert A. Bray's collaboration.

Top Co-Authors

Avatar

Howard M. Gebel

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alan Landay

Rush University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge