Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Am J Transplant ; 19(1): 21-31, 2019 01.
Article in English | MEDLINE | ID: mdl-29956477

ABSTRACT

The Lung session of the 2017 14th Banff Foundation for Allograft Pathology Conference, Barcelona focused on the multiple aspects of antibody-mediated rejection (AMR) in lung transplantation. Multidimensional approaches for AMR diagnosis, including classification, histological and immunohistochemical analysis, and donor- specific antibody (DSA) characterization with their current strengths and limitations were reviewed in view of recent research. The group also discussed the role of tissue gene expression analysis in the context of unmet needs in lung transplantation. The current best practice for monitoring of AMR and the therapeutic approach are summarized and highlighted in this report. The working group reached consensus of the major gaps in current knowledge and focused on the unanswered questions regarding pulmonary AMR. An important outcome of the meeting was agreement on the need for future collaborative research projects to address these gaps in the field of lung transplantation.


Subject(s)
Antibodies/immunology , Graft Rejection/immunology , Lung Transplantation , Lung/immunology , Allografts , Complement C4/immunology , Gene Expression Profiling , HLA Antigens/immunology , Humans , Immunohistochemistry , Isoantibodies/immunology , Peptide Fragments/immunology , Societies, Medical , Tissue Donors , Transplantation, Homologous
2.
Am J Transplant ; 18(5): 1151-1157, 2018 05.
Article in English | MEDLINE | ID: mdl-29316230

ABSTRACT

From a prospective and multicentric French cohort, we proposed an external validation study for the expanded criteria donor (ECD), based on 4833 kidney recipients transplanted for the first time between 2000 and 2014. We estimated the subject-specific effect from a multivariable Cox model. We confirmed a 1.75-fold (95% confidence interval [CI] 1.53-2.00, P < .0001) increase in graft failure risk if a given patient received an ECD graft compared to a graft from a donor with standard criteria (standard criteria donor [SCD]). Complementarily, we estimated the population-average effect using propensity scores. We estimated a 1.34-fold (95% CI 1.09-1.64, P = .0049) increase in graft failure risk among ECD patients receiving an ECD graft compared to receiving a SCD graft. With a 10-year follow-up, it corresponded to a decrease of 8 months of the mean time to graft failure due to ECD transplantation (95% CI 2-14 months). The population-average relative risk due to ECD transplantation and the corresponding absolute effect seem finally not so high. Regarding the increase of quality of life in transplantation, our study constitutes an argument to extend the definition of marginality by considering more grafts at high risk and thereby enlarging the pool of kidney grafts.


Subject(s)
Graft Rejection/mortality , Kidney Failure, Chronic/surgery , Kidney Transplantation/mortality , Propensity Score , Tissue Donors/supply & distribution , Tissue and Organ Procurement/methods , Tissue and Organ Procurement/statistics & numerical data , Adult , Aged , Donor Selection , Female , Follow-Up Studies , Graft Rejection/etiology , Graft Rejection/pathology , Graft Survival , Humans , Kidney Transplantation/adverse effects , Male , Middle Aged , Prognosis , Prospective Studies , Quality of Life , Risk Factors , Survival Rate , Time Factors , Tissue and Organ Procurement/standards , Transplant Recipients
3.
Am J Transplant ; 18 Suppl 3: 3-17, 2018 01.
Article in English | MEDLINE | ID: mdl-29292861

ABSTRACT

Despite the success of desensitization protocols, antibody-mediated rejection (AMR) remains a significant contributor to renal allograft failure in patients with donor-specific antibodies. Plasmapheresis and high-dose intravenous immunoglobulin have proved to be effective treatments to prevent and treat AMR, but irreversible injury in the form of transplant glomerulopathy can commonly manifest months to years later. There is an unmet need to improve the outcomes for patients at risk for AMR. Updated Banff criteria now take into account the increasing understanding of the complex and heterogeneous nature of AMR phenotypes, including the timing of rejection, subclinical and chronic AMR, C4d-negative AMR, and antibody-mediated vascular rejection. Treatment for AMR is not standardized, and there is little in the way of evidence-based treatment guidelines. Refining more precisely the mechanisms of injury responsible for different AMR phenotypes and establishing relevant surrogate endpoints to facilitate more informative studies will likely allow for more accurate determination of prognosis and efficacious intervention using new therapeutic approaches. In addition to plasma exchange and intravenous immunoglobulin, a number of other add-on therapies have been tried in small studies without consistent benefit, including anti-CD20, proteasome inhibitors, complement inhibitors, anti-interleukin-6 receptor blockers, and immunoglobulin G-degrading enzyme of Streptococcus pyogenes (called IdeS).


Subject(s)
Graft Rejection/etiology , Graft Rejection/prevention & control , Immunoglobulins, Intravenous/administration & dosage , Isoantibodies/adverse effects , Kidney Transplantation/adverse effects , Humans
4.
Am J Transplant ; 18(2): 293-307, 2018 02.
Article in English | MEDLINE | ID: mdl-29243394

ABSTRACT

The kidney sessions of the 2017 Banff Conference focused on 2 areas: clinical implications of inflammation in areas of interstitial fibrosis and tubular atrophy (i-IFTA) and its relationship to T cell-mediated rejection (TCMR), and the continued evolution of molecular diagnostics, particularly in the diagnosis of antibody-mediated rejection (ABMR). In confirmation of previous studies, it was independently demonstrated by 2 groups that i-IFTA is associated with reduced graft survival. Furthermore, these groups presented that i-IFTA, particularly when involving >25% of sclerotic cortex in association with tubulitis, is often a sequela of acute TCMR in association with underimmunosuppression. The classification was thus revised to include moderate i-IFTA plus moderate or severe tubulitis as diagnostic of chronic active TCMR. Other studies demonstrated that certain molecular classifiers improve diagnosis of ABMR beyond what is possible with histology, C4d, and detection of donor-specific antibodies (DSAs) and that both C4d and validated molecular assays can serve as potential alternatives and/or complements to DSAs in the diagnosis of ABMR. The Banff ABMR criteria are thus updated to include these alternatives. Finally, the present report paves the way for the Banff scheme to be part of an integrative approach for defining surrogate endpoints in next-generation clinical trials.


Subject(s)
Graft Rejection/diagnosis , High-Throughput Nucleotide Sequencing/methods , Inflammation/diagnosis , Isoantibodies/immunology , Kidney Transplantation/adverse effects , Postoperative Complications , T-Lymphocytes/immunology , Graft Rejection/etiology , Graft Rejection/pathology , Humans , Inflammation/etiology , Inflammation/pathology , Prognosis , Research Report
5.
Am J Transplant ; 17(11): 2851-2862, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28449409

ABSTRACT

The authors conducted a prospective trial to assess the feasibility of real time central molecular assessment of kidney transplant biopsy samples from 10 North American or European centers. Biopsy samples taken 1 day to 34 years posttransplantation were stabilized in RNAlater, sent via courier overnight at ambient temperature to the central laboratory, and processed (29 h workflow) using microarrays to assess T cell- and antibody-mediated rejection (TCMR and ABMR, respectively). Of 538 biopsy samples submitted, 519 (96%) were sufficient for microarray analysis (average length, 3 mm). Automated reports were generated without knowledge of histology and HLA antibody, with diagnoses assigned based on Molecular Microscope Diagnostic System (MMDx) classifier algorithms and signed out by one observer. Agreement between MMDx and histology (balanced accuracy) was 77% for TCMR, 77% for ABMR, and 76% for no rejection. A classification tree derived to provide automated sign-outs predicted the observer sign-outs with >90% accuracy. In 451 biopsy samples where feedback was obtained, clinicians indicated that MMDx more frequently agreed with clinical judgment (87%) than did histology (80%) (p = 0.0042). In 81% of feedback forms, clinicians reported that MMDx increased confidence in management compared with conventional assessment alone. The authors conclude that real time central molecular assessment is feasible and offers a useful new dimension in biopsy interpretation. ClinicalTrials.gov NCT#01299168.


Subject(s)
Biomarkers/metabolism , Gene Expression Profiling , Graft Rejection/diagnosis , Kidney Failure, Chronic/surgery , Kidney Transplantation/adverse effects , Adult , Aged , Aged, 80 and over , Biopsy , Female , Follow-Up Studies , Glomerular Filtration Rate , Graft Rejection/etiology , Graft Rejection/metabolism , Graft Survival , Humans , Kidney Function Tests , Male , Middle Aged , Prognosis , Prospective Studies , Risk Factors , Young Adult
6.
Am J Transplant ; 17(9): 2381-2389, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28199785

ABSTRACT

Extending the functional integrity of renal allografts is the primary goal of transplant medicine. The development of donor-specific antibodies (DSAs) posttransplantation leads to chronic active antibody-mediated rejection (cAMR) and transplant glomerulopathy (TG), resulting in the majority of graft losses that occur in the United States. This reduces the quality and length of life for patients and increases cost. There are no approved treatments for cAMR. Evidence suggests the proinflammatory cytokine interleukin 6 (IL-6) may play an important role in DSA generation and cAMR. We identified 36 renal transplant patients with cAMR plus DSAs and TG who failed standard of care treatment with IVIg plus rituximab with or without plasma exchange. Patients were offered rescue therapy with the anti-IL-6 receptor monoclonal tocilizumab with monthly infusions and monitored for DSAs and long-term outcomes. Tocilizumab-treated patients demonstrated graft survival and patient survival rates of 80% and 91% at 6 years, respectively. Significant reductions in DSAs and stabilization of renal function were seen at 2 years. No significant adverse events or severe adverse events were seen. Tocilizumab provides good long-term outcomes for patients with cAMR and TG, especially compared with historical published treatments. Inhibition of the IL-6-IL-6 receptor pathway may represent a novel approach to stabilize allograft function and extend patient lives.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Graft Rejection/drug therapy , HLA Antigens/immunology , Isoantibodies/adverse effects , Kidney Failure, Chronic/surgery , Kidney Transplantation/adverse effects , Receptors, Interleukin-6/antagonists & inhibitors , Female , Follow-Up Studies , Glomerular Filtration Rate , Graft Rejection/etiology , Graft Survival/drug effects , Graft Survival/immunology , Humans , Immunosuppressive Agents/therapeutic use , Kidney Function Tests , Male , Middle Aged , Prognosis , Receptors, Interleukin-6/immunology , Risk Factors , Transplantation, Homologous
7.
J Immunol Res ; 2017: 5201098, 2017.
Article in English | MEDLINE | ID: mdl-28133619

ABSTRACT

The purpose of the present review is to describe how we improve the model for risk stratification of transplant outcomes in kidney transplantation by incorporating the novel insights of donor-specific anti-HLA antibody (DSA) characteristics. The detection of anti-HLA DSA is widely used for the assessment of pre- and posttransplant risks of rejection and allograft loss; however, not all anti-HLA DSA carry the same risk for transplant outcomes. These antibodies have been shown to cause a wide spectrum of effects on allografts, ranging from the absence of injury to indolent or full-blown acute antibody-mediated rejection. Consequently, the presence of circulating anti-HLA DSA does not provide a sufficient level of accuracy for the risk stratification of allograft outcomes. Enhancing the predictive performance of anti-HLA DSA is currently one of the most pressing unmet needs for facilitating individualized treatment choices that may improve outcomes. Recent advancements in the assessment of anti-HLA DSA properties, including their strength, complement-binding capacity, and IgG subclass composition, significantly improved the risk stratification model to predict allograft injury and failure. Although risk stratification based on anti-HLA DSA properties appears promising, further specific studies that address immunological risk stratification in large and unselected populations are required to define the benefits and cost-effectiveness of such comprehensive assessment prior to clinical implementation.


Subject(s)
Allografts/immunology , Graft Rejection/immunology , HLA Antigens/immunology , Kidney Transplantation , Antibody Specificity , Female , Graft Rejection/prevention & control , Graft Survival/immunology , Humans , Isoantibodies/blood , Kidney Transplantation/adverse effects , Male , Risk Factors , Tissue Donors , Transplantation, Homologous
8.
Am J Transplant ; 17(1): 28-41, 2017 01.
Article in English | MEDLINE | ID: mdl-27862883

ABSTRACT

The XIII Banff meeting, held in conjunction the Canadian Society of Transplantation in Vancouver, Canada, reviewed the clinical impact of updates of C4d-negative antibody-mediated rejection (ABMR) from the 2013 meeting, reports from active Banff Working Groups, the relationships of donor-specific antibody tests (anti-HLA and non-HLA) with transplant histopathology, and questions of molecular transplant diagnostics. The use of transcriptome gene sets, their resultant diagnostic classifiers, or common key genes to supplement the diagnosis and classification of rejection requires further consensus agreement and validation in biopsies. Newly introduced concepts include the i-IFTA score, comprising inflammation within areas of fibrosis and atrophy and acceptance of transplant arteriolopathy within the descriptions of chronic active T cell-mediated rejection (TCMR) or chronic ABMR. The pattern of mixed TCMR and ABMR was increasingly recognized. This report also includes improved definitions of TCMR and ABMR in pancreas transplants with specification of vascular lesions and prospects for defining a vascularized composite allograft rejection classification. The goal of the Banff process is ongoing integration of advances in histologic, serologic, and molecular diagnostic techniques to produce a consensus-based reporting system that offers precise composite scores, accurate routine diagnostics, and applicability to next-generation clinical trials.


Subject(s)
Arteritis/immunology , Complement C4b/immunology , Graft Rejection/classification , Graft Rejection/pathology , Isoantibodies/immunology , Kidney Transplantation/adverse effects , Peptide Fragments/immunology , Graft Rejection/etiology , Humans , Research Report
9.
Am J Transplant ; 17(1): 42-53, 2017 01.
Article in English | MEDLINE | ID: mdl-27862968

ABSTRACT

The 13th Banff Conference on Allograft Pathology was held in Vancouver, British Columbia, Canada from October 5 to 10, 2015. The cardiac session was devoted to current diagnostic issues in heart transplantation with a focus on antibody-mediated rejection (AMR) and small vessel arteriopathy. Specific topics included the strengths and limitations of the current rejection grading system, the central role of microvascular injury in AMR and approaches to semiquantitative assessment of histopathologic and immunophenotypic indicators, the role of AMR in the development of cardiac allograft vasculopathy, the important role of serologic antibody detection in the management of transplant recipients, and the potential application of new molecular approaches to the elucidation of the pathophysiology of AMR and potential for improving the current diagnostic system. Herein we summarize the key points from the presentations, the comprehensive, open and wide-ranging multidisciplinary discussion that was generated, and considerations for future endeavors.


Subject(s)
Graft Rejection/pathology , Isoantibodies/immunology , Organ Transplantation/adverse effects , Practice Guidelines as Topic/standards , Graft Rejection/etiology , Humans , Isoantibodies/blood , Research Report , Transplantation, Homologous
10.
Am J Transplant ; 16(5): 1596-603, 2016 05.
Article in English | MEDLINE | ID: mdl-26693703

ABSTRACT

Complement inhibitors have not been thoroughly evaluated in the treatment of acute antibody-mediated rejection (ABMR). We performed a prospective, single-arm pilot study to investigate the potential effects and safety of C1 inhibitor (C1-INH) Berinert added to high-dose intravenous immunoglobulin (IVIG) for the treatment of acute ABMR that is nonresponsive to conventional therapy. Kidney recipients with nonresponsive active ABMR and acute allograft dysfunction were enrolled between April 2013 and July 2014 and received C1-INH and IVIG for 6 months (six patients). The primary end point was the change in eGFR at 6 months after inclusion (M+6). Secondary end points included the changes in histology and DSA characteristics and adverse events as evaluated at M+6. All patients showed an improvement in eGFR between inclusion and M+6: from 38.7 ± 17.9 to 45.2 ± 21.3 mL/min/1.73 m(2) (p = 0.0277). There was no change in histological features, except a decrease in the C4d deposition rate from 5/6 to 1/6 (p = 0.0455). There was a change in DSA C1q status from 6/6 to 1/6 positive (p = 0.0253). One deep venous thrombosis was observed. In a secondary analysis, C1-INH patients were compared with a similar historical control group (21 patients). C1-INH added to IVIG is safe and may improve allograft function in kidney recipients with nonresponsive acute ABMR.


Subject(s)
Complement C1 Inhibitor Protein/therapeutic use , Graft Rejection/drug therapy , Isoantibodies/immunology , Kidney Failure, Chronic/complications , Kidney Transplantation/adverse effects , Adult , Complement Inactivating Agents/therapeutic use , Female , Follow-Up Studies , Glomerular Filtration Rate , Graft Rejection/etiology , Graft Survival/drug effects , Humans , Immunoglobulins, Intravenous/administration & dosage , Kidney Failure, Chronic/surgery , Kidney Function Tests , Male , Middle Aged , Pilot Projects , Prognosis , Prospective Studies , Risk Factors
11.
Am J Transplant ; 16(1): 111-20, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26588356

ABSTRACT

In heart transplantation, there is a lack of robust evidence of the specific causes of late allograft failure. We hypothesized that a substantial fraction of failing heart allografts may be associated with antibody-mediated injury and immune-mediated coronary arteriosclerosis. We included all patients undergoing a retransplantation for late terminal heart allograft failure in three referral centers. We performed an integrative strategy of heart allograft phenotyping by assessing the heart vascular tree including histopathology and immunohistochemistry together with circulating donor-specific antibodies. The main analysis included 40 explanted heart allografts patients and 402 endomyocardial biopsies performed before allograft loss. Overall, antibody-mediated rejection was observed in 19 (47.5%) failing heart allografts including 16 patients (40%) in whom unrecognized previous episodes of subclinical antibody-mediated rejection occurred 4.5 ± 3.5 years before allograft loss. Explanted allografts with evidence of antibody-mediated rejection demonstrated higher endothelitis and microvascular inflammation scores (0.89 ± 0.26 and 2.25 ± 0.28, respectively) compared with explanted allografts without antibody-mediated rejection (0.42 ± 0.11 and 0.36 ± 0.09, p = 0.046 and p < 0.0001, respectively). Antibody-mediated injury was observed in 62.1% of failing allografts with pure coronary arteriosclerosis and mixed (arteriosclerosis and atherosclerosis) pattern, while it was not observed in patients with pure coronary atherosclerosis (p = 0.0076). We demonstrate that antibody-mediated rejection is operating in a substantial fraction of failing heart allografts and is associated with severe coronary arteriosclerosis. Unrecognized subclinical antibody-mediated rejection episodes may be observed years before allograft failure.


Subject(s)
Coronary Artery Disease/pathology , Graft Rejection/pathology , Heart Failure/surgery , Heart Transplantation/adverse effects , Isoantibodies/adverse effects , Adult , Allografts , Coronary Artery Disease/etiology , Female , Graft Rejection/etiology , Humans , Isoantibodies/blood , Male , Reoperation
13.
Am J Transplant ; 14(2): 272-83, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24472190

ABSTRACT

The 12th Banff Conference on Allograft Pathology was held in Comandatuba, Brazil, from August 19-23, 2013, and was preceded by a 2-day Latin American Symposium on Transplant Immunobiology and Immunopathology. The meeting was highlighted by the presentation of the findings of several working groups formed at the 2009 and 2011 Banff meetings to: (1) establish consensus criteria for diagnosing antibody-mediated rejection (ABMR) in the presence and absence of detectable C4d deposition; (2) develop consensus definitions and thresholds for glomerulitis (g score) and chronic glomerulopathy (cg score), associated with improved inter-observer agreement and correlation with clinical, molecular and serological data; (3) determine whether isolated lesions of intimal arteritis ("isolated v") represent acute rejection similar to intimal arteritis in the presence of tubulointerstitial inflammation; (4) compare different methodologies for evaluating interstitial fibrosis and for performing/evaluating implantation biopsies of renal allografts with regard to reproducibility and prediction of subsequent graft function; and (5) define clinically and prognostically significant morphologic criteria for subclassifying polyoma virus nephropathy. The key outcome of the 2013 conference is defining criteria for diagnosis of C4d-negative ABMR and respective modification of the Banff classification. In addition, three new Banff Working Groups were initiated.


Subject(s)
Arteritis/etiology , Complement C4b/metabolism , Graft Rejection/etiology , Isoantibodies/immunology , Organ Transplantation/adverse effects , Peptide Fragments/metabolism , Arteritis/metabolism , Graft Rejection/metabolism , Humans , Research Report
14.
Am J Transplant ; 13(4): 971-983, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23414212

ABSTRACT

Antibody-mediated rejection is the major cause of kidney transplant failure, but the histology-based diagnostic system misses most cases due to its requirement for C4d positivity. We hypothesized that gene expression data could be used to test biopsies for the presence of antibody-mediated rejection. To develop a molecular test, we prospectively assigned diagnoses, including C4d-negative antibody-mediated rejection, to 403 indication biopsies from 315 patients, based on histology (microcirculation lesions) and donor-specific HLA antibody. We then used microarray data to develop classifiers that assigned antibody-mediated rejection scores to each biopsy. The transcripts distinguishing antibody-mediated rejection from other conditions were mostly expressed in endothelial cells or NK cells, or were IFNG-inducible. The scores correlated with the presence of microcirculation lesions and donor-specific antibody. Of 45 biopsies with scores>0.5, 39 had been diagnosed as antibody-mediated rejection on the basis of histology and donor-specific antibody. High scores were also associated with unanimity among pathologists that antibody-mediated rejection was present. The molecular score also strongly predicted future graft loss in Cox regression analysis. We conclude that microarray assessment of gene expression can assign a probability of ABMR to transplant biopsies without knowledge of HLA antibody status, histology, or C4d staining, and predicts future failure.


Subject(s)
Antibodies/immunology , Graft Rejection/diagnosis , Graft Rejection/immunology , Kidney Transplantation , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Complement C4b/immunology , Endothelial Cells/cytology , Female , Gene Expression Regulation , Graft Survival , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/cytology , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Peptide Fragments/immunology , Probability , Proportional Hazards Models , Prospective Studies , Regression Analysis , Young Adult
16.
Am J Transplant ; 11(7): 1478-87, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21668629

ABSTRACT

In heart transplants, the significance of very late rejection (after 7 years post-transplant, VLR) detected by routine endomyocardial biopsies (EMB) remains uncertain. Here, we assessed the prevalence, histopathological and immunological phenotype, and outcome of VLR in clinically stable patients. Between 1985 and 2009, 10 662 protocol EMB were performed at our institution in 398 consecutive heart transplants recipients. Among the 196 patients with >7-year follow-up, 20 (10.2%) presented subclinical ≥3A/2R-ISHLT rejection. The VLR group was compared to a matched control group of patients without rejection. All biopsies were stained for C4d/C3d/CD68 with sera screened for the presence of donor-specific antibodies (DSAs). In addition to cellular infiltrates with myocyte damage, 60% of VLR patients had evidence of intravascular macrophages. C4d and/or C3d-capillary deposition was found in 55% VLR EMB. All cases of VLR associated with microcirculation injury had DSAs (mean DSA(max) -MFI = 1751 ± 583). This entity was absent from the control group (p < 0.0001). Finally, after a similar follow-up postreference EMB of 6.4 ± 1 years, the mean of CAV grade was 0.76 ± 0.18 in the control group compared to 2.06 ± 0.26 in the VLR group respectively, p = 0.001). There was no difference in patient survival between study and control groups. In conclusion, VLR is frequently associated with complement-cascade activation, microvascular injury and DSA, suggesting an antibody-mediated process. VLR is associated with a dramatic progression to severe CAV in long-term follow-up.


Subject(s)
Graft Rejection/immunology , Graft Rejection/pathology , Heart Transplantation/pathology , Adult , Antibodies/immunology , Complement Activation , Female , Follow-Up Studies , Heart Transplantation/immunology , Heart Transplantation/mortality , Humans , Male , Middle Aged , Retrospective Studies , Tissue Donors
17.
Am J Transplant ; 11(1): 56-65, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21199348

ABSTRACT

The significance of C4d-Banff scores in protocol biopsies of kidney transplant recipients with preformed donor-specific antibodies (DSA) has not been determined. We reviewed 157 protocol biopsies from 80 DSA+ patients obtained at 3 months and 1 year post-transplant. The C4d Banff scores (1,2,3) were associated with significant increments of microcirculation inflammation (MI) at both 3 months and 1 year post-transplant, worse transplant glomerulopathy and higher class II DSA-MFI (p < 0.01). Minimal-C4d had injury intermediate between negative and focal, while focal and diffuse-C4d had the same degree of microvascular injury. A total of 54% of patients had variation of C4d score between 3 months and 1 year post-transplant. Cumulative (3 month + 1 year) C4d scores correlated with long-term renal function worsening (p = 0.006). However, C4d staining was not a sensitive indicator of parenchymal disease, 55% of C4d-negative biopsies having evidence of concomitant MI. Multivariate analysis demonstrated that the presence of MI and class II DSA at 3 months were associated with a fourfold increased risk of progression to chronic antibody-mediated rejection independently of C4d (p < 0.05). In conclusion, the substantial fluctuation of C4d status in the first year post-transplant reflects a dynamic humoral process. However, C4d may not be a sufficiently sensitive indicator of activity, MI and DSA being more robust predictors of bad outcome.


Subject(s)
Complement C4b/immunology , Graft Rejection/immunology , Kidney Transplantation/immunology , Peptide Fragments/immunology , Adult , Aged , Antibodies , Biopsy , Graft Survival/immunology , Humans , Kidney/immunology , Kidney Transplantation/pathology , Microcirculation/immunology , Middle Aged
19.
Am J Transplant ; 10(3): 681-6, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20121729

ABSTRACT

Persistence of donor-specific anti-HLA antibodies (DSA) associated with antibody-mediated graft injuries following kidney transplantation predicts evolution toward chronic humoral rejection and reduced graft survival. Targeting plasma cells, the main antibody-producing cells, with the proteasome inhibitor bortezomib may be a promising desensitization strategy. We evaluated the in vivo efficacy of one cycle of bortezomib (1.3 mg/m(2)x 4 doses), used as the sole desensitization therapy, in four renal transplant recipients experiencing subacute antibody-mediated rejection with persisting DSA (>2000 [Mean Fluorescence Intensity] MFI). Bortezomib treatment did not significantly decrease DSA MFI within the 150-day posttreatment period in any patient. In addition, antivirus (HBV, VZV and HSV) antibody levels remained stable following treatment suggesting a lack of efficacy on long-lived plasma cells. In conclusion, one cycle of bortezomib alone does not decrease DSA levels in sensitized kidney transplant recipients in the time period studied. These results underscore the need to evaluate this new desensitization agent properly in prospective, randomized and well-controlled studies.


Subject(s)
Boronic Acids/therapeutic use , HLA Antigens/biosynthesis , Immunosuppressive Agents/therapeutic use , Kidney Transplantation/methods , Pyrazines/therapeutic use , Adult , Biopsy , Bortezomib , Female , Graft Survival , HLA Antigens/chemistry , Humans , Immunohistochemistry/methods , Male , Middle Aged , Randomized Controlled Trials as Topic , Time Factors , Treatment Outcome
20.
SELECTION OF CITATIONS
SEARCH DETAIL
...