Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Int J Immunogenet ; 47(3): 235-242, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32426916

ABSTRACT

Organ transplantation is an effective way to treat end-stage organ disease. Extending the graft survival is one of the major goals in the modern era of organ transplantation. However, long-term graft survival has not significantly improved in recent years despite the improvement of patient management and advancement of immunosuppression regimen. Antibody-mediated rejection is a major obstacle for long-term graft survival. Donor human leucocyte antigen (HLA)-specific antibodies were initially identified as a major cause for antibody-mediated rejection. Recently, with the development of solid-phase-based assay reagents, the contribution of non-HLA antibodies in organ transplantation starts to be appreciated. Here, we review the role of most studied non-HLA antibodies, including angiotensin II type 1 receptor (AT1 R), K-α-tubulin and vimentin antibodies, in the solid organ transplant, and discuss the possible mechanism by which these antibodies are stimulated.


Subject(s)
Antibodies/immunology , Graft Rejection/immunology , Receptor, Angiotensin, Type 1/immunology , Tubulin/immunology , Vimentin/immunology , Antibodies/genetics , Autoantibodies/immunology , Graft Rejection/genetics , Graft Survival/immunology , HLA Antigens/immunology , Humans , Organ Transplantation/adverse effects , Tissue Donors , Transplantation, Homologous/adverse effects
3.
Hum Immunol ; 80(8): 579-582, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30980862

ABSTRACT

Angiotensin II type I receptor (AT1R) is a critical player in regulating vasoconstriction, blood pressure, sodium retention. Even though AT1R has limited polymorphism, AT1R antibodies have been detected in several diseases. The role of AT1R antibodies in transplantation is first reported in kidney transplant, and then identified in heart and lung transplants. Mechanical circulatory support devices (MCS) can also stimulate production of AT1R antibodies. AT1R antibodies may negatively impact graft or patient survival through mechanisms independent of the classical complement activation.


Subject(s)
Autoantibodies/metabolism , Graft Rejection/immunology , Heart Transplantation , Lung Transplantation , Receptor, Angiotensin, Type 1/metabolism , Animals , Blood Circulation , Blood Pressure , Extracorporeal Circulation , Humans , Receptor, Angiotensin, Type 1/immunology , Sodium/metabolism , Thoracic Surgery , Vasoconstriction
4.
Hum Immunol ; 78(11-12): 699-703, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28899793

ABSTRACT

The single antigen test is widely used in the field of transplantation to determine the specificity of HLA antibodies. It will be beneficial to standardize the procedure of the single antigen test among HLA laboratories. It is not uncommon that single antigen testing on native sera fails to detect antibodies with very high concentrations. It has been shown that cleavage products of activated complement components may mask strongly binding antibodies in single antigen testing. To overcome inhibition by the activated complement products, sera are pretreated with ethylenediaminetetraacetic acid (EDTA), dithiothreitol (DTT), or heat inactivation before single antigen testing. However, no studies have been published to systemically compare the impact of these treatments on single antigen testing. The aim of this study is to understand the different effects these treatments may have on single antigen test results. We found that mean fluorescence intensity (MFI) obtained from sera treated with EDTA and heat inactivation were nearly identical, while DTT treatment was less potent to remove the inhibition. In addition, sera dilution did not further increase MFI of antibodies after EDTA treatment. Our results provide guidance to choose a pretreatment reagent for single antigen testing, and to compare studies obtained from laboratories using different treatments.


Subject(s)
Graft Rejection/immunology , Histocompatibility Testing/methods , Isoantibodies/metabolism , Kidney Transplantation , Complement System Proteins/metabolism , Dithiothreitol/metabolism , Edetic Acid/metabolism , Epitopes/immunology , HLA Antigens/immunology , Hot Temperature , Humans , Immunity, Humoral
5.
Front Immunol ; 8: 434, 2017.
Article in English | MEDLINE | ID: mdl-28450866

ABSTRACT

The presence of donor human leukocyte antigen (HLA)-specific antibodies has been shown to be associated with graft loss and decreased patient survival, but it is not uncommon that donor-specific HLA antibodies are absent in patients with biopsy-proven antibody-mediated rejection. In this review, we focus on the latest findings on antibodies against non-HLA antigens in kidney and heart transplantation. These non-HLA antigens include myosin, vimentin, Kα1 tubulin, collagen, and angiotensin II type 1 receptor. It is suggested that the detrimental effects of HLA antibodies and non-HLA antibodies synergize together to impact graft outcome. Injury of graft by HLA antibodies can cause the exposure of neo-antigens which in turn stimulate the production of antibodies against non-HLA antigens. On the other hand, the presence of non-HLA antibodies may increase the risk for a patient to develop HLA-specific antibodies. These findings indicate it is imperative to stratify the patient's immunologic risk by assessing both HLA and non-HLA antibodies.

6.
Kidney Int ; 91(3): 729-737, 2017 03.
Article in English | MEDLINE | ID: mdl-28104301

ABSTRACT

Antibody-mediated rejection (ABMR) of renal allografts occurs in two forms. Type 1 ABMR results from persistence and/or a rebound of preexisting donor-specific antibodies in sensitized patients and usually occurs early post-transplantation. Type 2 ABMR is associated with de novo donor-specific antibodies and usually occurs over one year post-transplantation. It is generally accepted that types 1 and 2 also differ with regard to certain pathologic features including the frequencies of C4d positivity and concurrent cell-mediated rejection. However, direct comparison of pathologic, serologic, and clinical features of types 1 and 2 ABMR is lacking. Here we compared these features in 80 cases of ABMR (37 type 1, 43 type 2) diagnosed at our center. Compared with type 1, type 2 ABMR occurred later post-transplantation, was more often associated with donor-specific antibodies against Class II HLA, and was associated with more interstitial fibrosis/tubular atrophy and more frequent cell-mediated rejection, although these did not differ with respect to C4d positivity. By univariate analysis, graft survival was lower with type 2 than type 1 ABMR with borderline significance. Still, among these 80 patients, all but one treated for ABMR following diagnosis, the only two independent predictors of graft failure were at least moderate interstitial fibrosis/tubular atrophy and failure of the donor-specific antibody relative intensity scale score, a measure of the combined strength of all donor-specific antibodies present, to decrease in response to therapy.


Subject(s)
Graft Rejection/diagnosis , Graft Survival , HLA Antigens/immunology , Isoantibodies/blood , Kidney Transplantation/adverse effects , Kidney/immunology , Kidney/pathology , Adult , Allografts , Atrophy , Biopsy , Complement C4b/analysis , Female , Fibrosis , Graft Rejection/drug therapy , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival/drug effects , Histocompatibility , Humans , Immunosuppressive Agents/therapeutic use , Kaplan-Meier Estimate , Kidney/drug effects , Los Angeles , Male , Middle Aged , Peptide Fragments/analysis , Predictive Value of Tests , Proportional Hazards Models , Retrospective Studies , Risk Factors , Serologic Tests , Time Factors
7.
Transplantation ; 101(6): 1215-1221, 2017 06.
Article in English | MEDLINE | ID: mdl-27973391

ABSTRACT

BACKGROUND: The presence of antibodies to angiotensin type 1 receptor (AT1R) and endothelin type A receptor (ETAR) is associated with allograft rejection in kidney and heart transplantation. The aim of our study was to determine the impact of AT1R and ETAR antibodies on graft outcome in lung transplantation. METHODS: Pretransplant and posttransplant sera from 162 lung recipients transplanted at 3 centers between 2011 and 2013 were tested for antibodies to AT1R and ETAR by the enzyme-linked immunosorbent assay (ELISA) assay. Clinical parameters analyzed were: HLA antibodies at transplant, de novo donor-specific antibodies (DSA), antibody-mediated rejection (AMR), acute cellular rejection, and graft status. RESULTS: Late AMR (median posttransplant day 323) was diagnosed in 5 of 36 recipients with de novo DSA. Freedom from AMR significantly decreased for those recipients with strong/intermediate binding antibodies to AT1R (P = 0.014) and ETAR (P = 0.005). Trends for lower freedom from acute cellular rejection were observed for recipients with pretransplant antibodies to AT1R (P = 0.19) and ETAR (P = 0.32), but did not reach statistical significance. Lower freedom from the development of de novo DSA was observed for recipients with antibodies detected pretransplant to AT1R (P = 0.054), ETAR (P = 0.012), and HLA-specific antibodies (P = 0.063). When the pretransplant antibody status of HLA-specific antibody (hazard ratio [HR], 1.69) was considered together with either strong binding to AT1R or ETAR, an increased negative impact on the freedom from the development of de novo DSA was observed (HR, 2.26 for HLA antibodies and ETAR; HR, 2.38 for HLA antibodies and ETAR). CONCLUSIONS: These results illustrate the increased negative impact when antibodies to both HLA and non-HLA antigens are present pretransplant.


Subject(s)
Graft Rejection/immunology , Histocompatibility , Isoantibodies/immunology , Lung Transplantation/adverse effects , Receptor, Angiotensin, Type 1/immunology , Receptor, Endothelin A/immunology , Disease-Free Survival , Enzyme-Linked Immunosorbent Assay , Graft Rejection/blood , Graft Survival , HLA Antigens/immunology , Histocompatibility Testing/methods , Humans , Isoantibodies/blood , Kaplan-Meier Estimate , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome , United States
9.
Curr Opin Organ Transplant ; 21(4): 350-4, 2016 08.
Article in English | MEDLINE | ID: mdl-27386832

ABSTRACT

PURPOSE OF REVIEW: Development of donor human leukocyte antigen (HLA)-specific antibodies is associated with graft loss, yet the role of non-HLA antibodies in solid organ transplant needs to be further defined. It is suggested that HLA antibodies and non-HLA antibodies collaborate together to impact graft outcome. This review focuses on the latest findings on antibodies against these non-HLA antigens in thoracic organ transplant. RECENT FINDINGS: These non-HLA antigens include signaling proteins expressed on the cell surface, such as angiotensin II type 1 receptor (AT1R), endothelin type A receptor, and structure proteins, such as myosin, vimentin, and Kα1 tubulin, and extracellular matrix protein collagen. Antibodies against these antigens may impact the allograft in different ways. Although these non-HLA antibodies can damage the allograft through complement-mediated or cell-mediated cytotoxicity, antibodies against AT1R and endothelin type A receptor can also alter the endothelial cell function by activating intracellular signals. The presence of these non-HLA antibodies may predispose the patient to develop HLA-specific antibodies. Recently, it has been shown patients with AT1R antibodies pretransplant have a higher chance to develop de-novo donor-specific HLA antibodies. SUMMARY: The findings suggest it is important to stratify the patient's immunologic risk by assessing both the HLA and non-HLA-specific antibodies.


Subject(s)
HLA Antigens/immunology , Thoracic Surgical Procedures/methods , Transplantation, Homologous/methods , Antibodies , Histocompatibility Antigens Class II , Humans , Tissue Donors
10.
J Heart Lung Transplant ; 35(4): 397-406, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27044531

ABSTRACT

Antibody-mediated rejection (AMR) is a recognized cause of allograft dysfunction in lung transplant recipients. Unlike AMR in other solid-organ transplant recipients, there are no standardized diagnostic criteria or an agreed-upon definition. Hence, a working group was created by the International Society for Heart and Lung Transplantation with the aim of determining criteria for pulmonary AMR and establishing a definition. Diagnostic criteria and a working consensus definition were established. Key diagnostic criteria include the presence of antibodies directed toward donor human leukocyte antigens and characteristic lung histology with or without evidence of complement 4d within the graft. Exclusion of other causes of allograft dysfunction increases confidence in the diagnosis but is not essential. Pulmonary AMR may be clinical (allograft dysfunction which can be asymptomatic) or sub-clinical (normal allograft function). This consensus definition will have clinical, therapeutic and research implications.


Subject(s)
Consensus , Graft Rejection/immunology , Heart Transplantation , International Cooperation , Lung Transplantation , Societies, Medical , Humans , Isoantibodies/immunology , Tissue Donors , Transplantation, Homologous
11.
J Heart Lung Transplant ; 35(2): 165-72, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26683810

ABSTRACT

BACKGROUND: The virtual crossmatch relies on the assignment of unacceptable antigens (UAs) to identify compatible donors. The purpose of our study was to identify an algorithm for assignment of UAs such that a negative complement-dependent cytotoxicity (CDC) crossmatch and concomitant negative or weakly positive flow cytometric crossmatch (FXM) are obtained. METHODS: We used 4 antibody methods: (1) Luminex single antigen (LSA), (2) LSA with a 1:8 serum dilution, (3) C1q LSA, and (4) CDC panel. The UAs were prioritized in the following order: (1) all C1q+/CDC+, (2) LSA 1:8 >7,500 median fluorescence intensity, and (3) LSA >10,000 median fluorescence intensity. RESULTS: Of 295 heart transplants that were performed at our center, 69 (23%) recipients had detectable human leukocyte antigen specific antibody at the time of transplant. All donor specific antibodies (DSAs) were avoided for 44 of 69 (64%) (DSA-). There were 25 recipients who had DSA at the time of transplant: 12 (48%) had negative FXM (DSA+/FXM-), and 13 (52%) had positive T-cell and/or B-cell FXM (DSA+/FXM+). Lower freedom from antibody-mediated rejection was observed for the DSA+/FXM+ group compared with the DSA- group (p < 0.0001). DSA remained detectable after transplant in the sera of 14 recipients, and de novo DSA was detected in 32 recipients. Freedom from antibody-mediated rejection was comparable for both groups (p = 0.53) but was lower than the DSA- group (p < 0.0001). Survival was comparable for all groups at 1,200 days post-transplant. CONCLUSIONS: Strategic prioritization of UA assignment has allowed transplantation of highly sensitized patients across the DSA barrier with survival rates comparable to DSA- heart transplant recipients.


Subject(s)
HLA Antigens/analysis , Heart Transplantation , Myocardium/immunology , Algorithms , Desensitization, Immunologic , Female , Flow Cytometry , Histocompatibility Testing , Humans , Immunosuppression Therapy , Isoantibodies/analysis , Male , Middle Aged , Postoperative Care , Tissue Donors
12.
Transplantation ; 97(5): 595-601, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24162250

ABSTRACT

BACKGROUND: De novo donor HLA-specific (dnDSA) and non-HLA antibodies including antiangiotensin type 1 receptor antibodies (AT1R-abs) have been associated with antibody-mediated rejection (AMR) and decreased graft survival as well as cellular-mediated rejection (CMR) and early onset of microvasculopathy in heart transplantation. The aim of our study was to determine the impact of anti-AT1R-ab and anti-donor HLA-specific antibody (DSA) on clinical outcomes. METHODS: Pretransplant and posttransplant sera from 200 recipients transplanted between May 2007 and August 2011 were tested for DSA (Luminex-based single antigen bead assay) and AT1R-ab (enzyme-linked immunosorbent assay). Two cutoff levels (≥ 17 and ≥ 12 units) were used to define high and intermediate binding of AT1R-ab. Clinical parameters examined were 5-year AMR/CMR (≥ grade 2), coronary artery vasculopathy, and survival. RESULTS: At 2 years after transplant, freedom from AMR and/or CMR was 95.4% for those with no DSA (n=175), 66.9% for those with dnDSA (n=19), and 25% for those with DSA at transplant (n=6) (P<0.0001). Neither ≥ 17 nor ≥ 12 units of pretransplant levels indicated a significant difference in freedom from AMR and/or CMR. When both dnDSA and AT1R-ab ≥ 17 or ≥ 12 units were considered, freedom from AMR and/or CMR decreased to 50% and 45% (P<0.0001), respectively. Coronary artery vasculopathy and survival were not significantly impacted. CONCLUSIONS: These results show the increased negative impact of dnDSA and AT1R-ab on freedom from AMR and/or CMR and an increased hazard ratio when both parameters are considered. Both HLA- and non-HLA-specific antibodies seem to impact graft outcome in heart transplantation.


Subject(s)
Antibodies/immunology , Graft Rejection/immunology , HLA Antigens/immunology , Heart Transplantation , Receptor, Angiotensin, Type 1/immunology , Tissue Donors , Antibodies/blood , Antibody Specificity/immunology , Follow-Up Studies , Graft Rejection/blood , Graft Rejection/epidemiology , Histocompatibility/immunology , Humans , Immune Tolerance/immunology , Incidence , Kaplan-Meier Estimate , Retrospective Studies , Transplantation
13.
Hum Immunol ; 74(11): 1474-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23831255

ABSTRACT

Angiotensin type I receptor (AT1R) mediates physiologic and pathophysiologic actions of its ligand, angiotensin II. Overactivity of the AT1R and angiotensin II interaction results in hypertension and vascular remodeling. Antibodies to AT1R have been implicated in several vascular pathologies. In renal transplantation, elevated levels of anti-AT1R antibodies have been associated with antibody mediated rejection (AMR) in the absence of donor HLA specific antibodies. In heart transplantation, increased levels of anti-AT1R antibodies have been associated with cellular and AMR as well as an early onset of microvasculopathy. This review summarizes the current investigations regarding the impact of anti-AT1R antibodies in solid organ transplantation and provides insight into the humoral response in the presence of non-HLA and HLA specific antibodies.


Subject(s)
Angiotensin II/immunology , Antibodies/immunology , Hypertension/immunology , Organ Transplantation , Receptor, Angiotensin, Type 2/immunology , Antibodies/blood , HLA Antigens/immunology , Humans , Transplantation Immunology
14.
Methods Mol Biol ; 1034: 353-8, 2013.
Article in English | MEDLINE | ID: mdl-23775750

ABSTRACT

The intracellular cytokine method allows for a multiparametric readout by flow cytometry with precise phenotyping of the responding T cells. The intracellular cytokine staining of cells that have been fixed and permeabilized following a short-term activation and staining with antibodies to cell surface markers allows for identification of the cellular origin of the cytokine accumulated product.


Subject(s)
Antibodies/immunology , Cytokines/isolation & purification , Lymphocyte Activation , T-Lymphocytes/immunology , Cell Membrane/immunology , Cell Membrane Permeability/immunology , Cytokines/immunology , Cytoplasm/immunology , Flow Cytometry , Humans , Protein Transport/immunology
15.
Chest ; 144(1): 226-233, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23328795

ABSTRACT

BACKGROUND: Long-term survival after lung transplant is limited by the development of chronic and progressive airflow obstruction, a condition known as bronchiolitis obliterans syndrome (BOS). While prior studies strongly implicate cellular rejection as a strong risk factor for BOS, less is known about the clinical significance of human leukocyte antigen (HLA) antibodies and donor HLA-specific antibodies in long-term outcomes. METHODS: A single-center cohort of 441 lung transplant recipients, spanning a 10-year period, was prospectively screened for HLA antibodies after transplant using flow cytometry-based methods. The prevalence of and predictors for HLA antibodies were determined. The impact of HLA antibodies on survival after transplant and the development of BOS were determined using Cox models. RESULTS: Of the 441 recipients, 139 (32%) had detectable antibodies to HLA. Of these 139, 54 (39%) developed antibodies specific to donor HLA. The detection of posttransplant HLA antibodies was associated with BOS (HR, 1.54; P=.04) and death (HR, 1.53; P=.02) in multivariable models. The detection of donor-specific HLA antibodies was associated with death (HR, 2.42; P<.0001). The detection of posttransplant HLA antibodies was associated with pretransplant HLA-antibody detection, platelet transfusions, and the development of BOS and cytomegalovirus pneumonitis. CONCLUSIONS: Approximately one-third of lung transplant recipients have detectable HLA antibodies, which are associated with a worse prognosis regarding graft function and patient survival.


Subject(s)
Antibodies/blood , Bronchiolitis Obliterans/epidemiology , Bronchiolitis Obliterans/mortality , HLA Antigens/immunology , Lung Transplantation/immunology , Adult , Biomarkers/blood , Cohort Studies , Female , Follow-Up Studies , Humans , Longitudinal Studies , Male , Middle Aged , Prevalence , Prognosis , Proportional Hazards Models , Retrospective Studies , Risk Factors , Survival Rate
16.
Eur Heart J ; 34(1): 68-75, 2013 Jan.
Article in English | MEDLINE | ID: mdl-21659438

ABSTRACT

AIMS: Cardiosphere-derived cells (CDCs) are in clinical development as a regenerative cell product which can be expanded ex vivo from patient cardiac biopsies. Cardiosphere-derived cells are clonogenic, exhibit multilineage differentiation, and exert functional benefits in preclinical models of heart failure. The origin of CDCs remains unclear: are these cells endogenous to the heart, or do they arise from cells that populate the heart via blood-borne seeding? METHODS AND RESULTS: Right ventricular endomyocardial biopsies were obtained from cardiac transplant recipients (n = 10, age 57 ± 15 years), and CDCs expanded from each biopsy. Donor-recipient mismatches were used to probe the origin of CDCs in three complementary ways. First, DNA analysis of short-tandem nucleotide repeats (STRs) was performed on genomic DNA from donor and recipient, then compared with the STR pattern of CDCs. Second, in two cases where the donor was male and the recipient female, CDCs were examined for the presence of X and Y chromosomes by fluorescence in situ hybridization. Finally, in two cases, quantitative PCR (qPCR) was performed for individual-specific polymorphisms of a major histocompatability locus to quantify the contribution of recipient cells to CDCs. In no case was recipient DNA detectable in the CDCs by STR analysis. In the two cases in which a female patient had received a male heart, all CDCs examined had an X and Y chromosome, similarly indicating exclusively donor origin. Likewise, qPCR on CDCs did not detect any recipient DNA. CONCLUSION: Cardiosphere-derived cells are of endogenous cardiac origin, with no detectable contribution from extra-cardiac seeding.


Subject(s)
Heart Ventricles/cytology , Myocardium/cytology , Myocytes, Cardiac/cytology , Stem Cells/cytology , Adult , Aged , Cell Differentiation/physiology , Cells, Cultured , DNA/analysis , Female , Heart Transplantation , Humans , In Situ Hybridization, Fluorescence , Male , Microsatellite Repeats , Middle Aged , Real-Time Polymerase Chain Reaction , Stem Cell Transplantation/methods , Young Adult
17.
Clin Transpl ; : 393-8, 2013.
Article in English | MEDLINE | ID: mdl-25095534

ABSTRACT

The implementation of the solid phase antibody assays has allowed for the detection and characterization of human leukocyte antigen (HLA) specific antibodies with greater sensitivity and specificity. This information can then be used along with the donor's HLA typing to predict crossmatch results (a virtual crossmatch). Using these data and the level of immunological risk assessed to the antibodies detected, the determination of unacceptable antigens can be made. The calculated panel reactive antibody (CPRA) provides for a means to determine the frequency of these unacceptable antigens in the donor population and thereby predict the probability of a positive crossmatch. In 2009, the Organ Procurement Transplant Network administered by the United Network for Organ Sharing adopted the CPRA as the means to define sensitization and to assign allocation points. Follow-up studies have shown that the number of organ offers refused due to a positive crossmatch has decreased significantly and has saved money through the elimination of unnecessary testing. An additional benefit has been the increased number of sensitized patients being transplanted successfully. Through technical improvements and the refined interpretation of the solid phase antibody assays, continual progress is being made in the definition of the unacceptable antigens and the ability to transplant sensitized patients.


Subject(s)
HLA Antigens/immunology , Histocompatibility Testing/methods , Organ Transplantation , Tissue and Organ Procurement , Enzyme-Linked Immunosorbent Assay/methods , Enzyme-Linked Immunosorbent Assay/trends , Flow Cytometry/methods , Flow Cytometry/trends , Histocompatibility Testing/trends , Humans , Resource Allocation , Sensitivity and Specificity , Tissue Donors , United States
19.
Discov Med ; 13(71): 267-73, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22541614

ABSTRACT

The broadly human leukocyte antigen (HLA) sensitized patient awaiting organ transplantation remains a persistent and significant problem for transplant medicine. Sensitization occurs as a consequence of exposure to HLA antigens through pregnancy, blood and platelet transfusions, and previous transplants. Early experience with desensitization protocols coupled with improved diagnostics for donor-specific antibodies (DSAs) and renal pathology have greatly improved transplant rates and outcomes for patients once considered un-transplantable or at high risk for poor outcomes. More recent advances have occurred through implementation of a national allocation system requiring the entering of unacceptable antigens that reduces the rate of crossmatch positivity. Current desensitization therapies include high-dose intravenous immunoglobulin (IVIG), plasma exchange (PLEX) with low-dose IVIG, and IVIG combined with rituximab. Developing therapies include proteasome inhibitors aimed at plasma cells and modifiers of complement-mediated injury. Here we discuss the important advancements in desensitization including defining the risk for antibody-mediated rejection prior to transplantation and the evolution of therapies aimed at reducing the impact of antibody injury on allografts.


Subject(s)
Desensitization, Immunologic/methods , HLA Antigens/immunology , Adult , Female , Humans , Immunoglobulins, Intravenous/therapeutic use , Kidney Transplantation/immunology
20.
Ann Thorac Surg ; 92(6): 2104-10; discussion 2111, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22115224

ABSTRACT

BACKGROUND: Heart transplantation for sensitized patients has been a significant challenge. In this study, outcome of heart transplantation in sensitized patients with virtual cross match was compared with prospective cross match. METHODS: Prior to July 2007, prospective cross match was used and afterward, virtual cross match with Luminex (One Lambda, Inc, Canoga Park, CA) based antibody analysis was used for potential heart transplant recipients. Prospectively collected data for the 3 years before and after July 2007, in sensitized (panel reactive antibody greater than 10%) and nonsensitized heart transplant recipients were reviewed. RESULTS: One hundred sixty-eight patients met inclusion criteria for analysis (78 patients for prospective cross match and 90 patients for virtual cross match). Multiple parameters were compared for the prospective cross match and virtual cross match eras. Three-year survivals in nonsensitized patients were 84.6% and 77.2% and in sensitized patients were 76.9% and 77.4% (p = 0.49) for prospective cross match and virtual cross match eras, respectively. Freedom from 3A (2R) cellular rejection in nonsensitized patients was 96.9% and 95.3%, and in sensitized patients was 90.9% and 100% (p = 0.83). Freedom from antibody-mediated rejection in nonsensitized patients was 95.3% and 96.8%, and in sensitized patients was 90.9% and 90.5% (p = 0.65). Mean waiting time was 129 ± 246 days (mean ± SD) for the period before virtual cross match and 59 ± 78 days with virtual cross match (p = 0.018). Donor geographic area was similar for prospective and virtual cross match. CONCLUSIONS: In sensitized heart transplant candidates, virtual cross match may shorten waiting time to heart transplantation without increasing subsequent occurrence of cellular rejection, antibody mediated rejection, and mortality after heart transplantation.


Subject(s)
Heart Transplantation , Histocompatibility Testing , Adult , Aged , Female , Graft Rejection , Heart Transplantation/mortality , Humans , Male , Middle Aged , Prospective Studies , Time Factors , Waiting Lists
SELECTION OF CITATIONS
SEARCH DETAIL
...