100 research outputs found

    Facteurs prédictifs de récidive et de sévérité du syndrome hémolytique et urémique atypique et de la glomérulonéphrite à C3 aprÚs transplantation rénale

    Get PDF
    Pas de rĂ©sumĂ© en anglaisLe syndrome hĂ©molytique et urĂ©mique atypique (SHU) et les glomĂ©rulonĂ©phrites Ă  dĂ©pĂŽts de C3 sont des pathologies rares. Le SHU atypique est une pathologie aigĂŒe sĂ©vĂšre, responsable de microangiopathie thrombotique rĂ©nale glomĂ©rulaire et artĂ©riolaire. La glomĂ©rulonĂ©phrite Ă  dĂ©pĂŽts de C3 (GNC3) est une pathologie chronique responsable d’une protĂ©inurie et d’une insuffisance rĂ©nale chronique qui Ă©volue sur plusieurs annĂ©es. Ces pathologies sont secondaires soit Ă  un dĂ©faut de rĂ©gulation soit Ă  une hyperactivation de la voie alterne du complĂ©ment. Dans ces deux pathologies, le risque de rĂ©cidive aprĂšs transplantation rĂ©nale est Ă©levĂ© et responsable d’une perte de greffon. L’objectif de ma thĂšse a Ă©tĂ© de rechercher des marqueurs prĂ©dictifs de rĂ©cidive et de sĂ©vĂ©ritĂ© conditionnant le pronostic de la transplantation rĂ©nale pour ces deux pathologies complĂ©ment–dĂ©pendantes. Une approche Ă  la fois clinique, biologique et gĂ©nĂ©tique a permis de dĂ©terminer pour le SHU atypique : les facteurs de risque gĂ©nĂ©tique de rĂ©cidive, de dĂ©terminer pour la premiĂšre fois que la prĂ©sence de l’haplotype homozygote du Facteur H (gtgt) est associĂ©e Ă  risque de rĂ©cidive et de prĂ©ciser les consĂ©quences fonctionnelles des mutations. Ce travail a contribuĂ© Ă  Ă©tablir les recommandations du traitement chez les patients transplantĂ©s pour un SHU atypique. Les facteurs de risque de rĂ©cidives des glomĂ©rulonĂ©phrites Ă  C3 ont Ă©tĂ© recherchĂ©s Ă  partir d’une large cohorte de patients que j’ai crĂ©Ă©e. Ce travail a permis de mettre en Ă©vidence une corrĂ©lation entre l’hyperactivation de la voie alterne du complĂ©ment (Ă©lĂ©vation du C5b9 soluble, C3 plasmatique bas), la prĂ©sence de dĂ©pĂŽts (C3, C5b9) dans le rein en post transplantation et la sĂ©vĂ©ritĂ© de la rĂ©cidive. La consommation de C3 en phase fluide, la prĂ©sence d’un sC5b9 Ă©levĂ© et la prĂ©sence de C5b9 in situ sont associĂ©es Ă  une rĂ©cidive sĂ©vĂšre. Enfin, j’ai participĂ© Ă  la premiĂšre description de mutations de DGKE dans le SHU atypique. J’ai mis au point la technique d’immunohistochimie permettant de dĂ©tecter et localiser DGKE dans les cellules endothĂ©liales et podocytaires rĂ©nales. Ce travail a permis de montrer que le DGKE est prĂ©sent dans l’endothĂ©lium rĂ©nal, Ă©lĂ©ment essentiel du mĂ©canisme physiopathologique du SHU atypique. Le DGKE est apportĂ© par le rein transplantĂ© et permet de corriger le risque de rĂ©cidive aprĂšs transplantation chez les patients dĂ©ficitaires

    Efficacy and Safety of Iptacopan in Patients With C3 Glomerulopathy

    Get PDF
    Complement 3 glomerulopathy; Inflammatory kidney disease; IptacopanGlomerulopatia del complement 3; Malaltia inflamatĂČria del ronyĂł; IptacopanGlomerulopatĂ­a del complemento 3; Enfermedad inflamatoria del riñón; IptacopanIntroduction Complement 3 glomerulopathy (C3G) is a rare inflammatory kidney disease mediated by dysregulation of the alternative complement pathway. No targeted therapy exists for this aggressive glomerulonephritis. Efficacy, safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) (measured by complement biomarkers) of iptacopan were assessed in patients with C3G. Methods In this phase 2, multicenter, open-label, single-arm, nonrandomized study, adults with biopsy-proven, native kidney C3G (native cohort) and kidney transplant recipients with C3G recurrence (recurrent kidney transplant [KT] cohort) received iptacopan twice daily (bid) for 84 days (days 1–21: 10–100 mg; days 22–84: 200 mg). The primary end point was the urine protein-to-creatinine ratio (UPCR; native cohort) and the change in the C3 deposit score of kidney biopsy (recurrent KT cohort). The complement pathway measures included Wieslab assay, soluble C5b9, and serum C3 levels. Results A total of 27 patients (16 native cohort and 11 recurrent KT cohort) were enrolled and all completed the study. In the native cohort, UPCR levels decreased by 45% from baseline to week 12 (P = 0.0003). In the recurrent KT cohort, the median C3 deposit score decreased by 2.50 (scale: 0–12) on day 84 versus baseline (P = 0.03). Serum C3 levels were normalized in most patients; complement hyperactivity observed pretreatment was reduced. Severe adverse events (AEs) included post-biopsy hematuria and hyperkalemia. No deaths occurred during the study. Conclusion Iptacopan resulted in statistically significant and clinically important reductions in UPCR and normalization of serum C3 levels in the native cohort and reduced C3 deposit scores in the recurrent KT cohort with favorable safety and tolerability. (ClinicalTrials.gov identifier: NCT03832114).The study was funded by Novartis Pharma AG, Basel, Switzerland

    Combined Liver-Kidney Transplantation With Preformed Anti-human Leukocyte Antigen Donor-Specific Antibodies

    Get PDF
    Introduction: the impact of preformed donor-specific anti-human leukocyte antigen (HLA) antibodies (pDSAs) after combined liver-kidney transplantation (CLKT) is still uncertain. Methods: we conducted a retrospective study in 8 European high-volume transplant centers and investigated the outcome of 166 consecutive CLKTs, including 46 patients with pDSAs. Results: patient survival was lower in those with pDSAs (5-year patient survival rate of 63% and 78% with or without pDSA, respectively; P = 0.04). The presence of pDSAs with a mean fluorescence intensity (MFI) ≄ 5000 (hazard ratio 4.96; 95% confidence interval: 2.3-10.9; P < 0.001) and the presence of 3 or more pDSAs (hazard ratio 6.5; 95% confidence interval: 2.5-18.8; P = 0.05) were independently associated with death. The death-censored liver graft survival was similar in patients with or without pDSAs. Kidney graft survival was comparable in both groups. (The 1- and 5-year death-censored graft survival rates were 91.6% and 79.5%, respectively, in patients with pDSAs and 93% and 88%, respectively, in the donor-specific antibody [DSA]-negative group, P = not significant). Despite a higher rate of kidney graft rejection in patients with pDSAs (5-year kidney graft survival rate without rejection of 87% and 97% with or without pDSAs, respectively; P = 0.04), kidney function did not statistically differ between both groups at 5 years post-transplantation (estimated glomerular filtration rate 45 ± 17 vs. 57 ± 29 ml/min per 1.73 m2, respectively, in patients with and without pDSAs). Five recipients with pDSAs (11.0%) experienced an antibody-mediated kidney rejection that led to graft loss in 1 patient. Conclusion: our results suggest that CLKT with pDSAs is associated with a lower patients' survival despite good recipients', liver and kidney grafts' outcome

    C5b9 Deposition in Glomerular Capillaries Is Associated With Poor Kidney Allograft Survival in Antibody-Mediated Rejection

    Get PDF
    C4d deposition in peritubular capillaries (PTC) reflects complement activation in antibody-mediated rejection (ABMR) of kidney allograft. However, its association with allograft survival is controversial. We hypothesized that capillary deposition of C5b9—indicative of complement-mediated injury—is a severity marker of ABMR. This pilot study aimed to determine the frequency, location and prognostic impact of these deposits in ABMR. We retrospectively selected patients diagnosed with ABMR in two French transplantation centers from January 2005 to December 2014 and performed C4d and C5b9 staining by immunohistochemistry. Fifty-four patients were included. Median follow-up was 52.5 (34.25–73.5) months. Thirteen patients (24%) had C5b9 deposits along glomerular capillaries (GC). Among these, seven (54%) had a global and diffuse staining pattern. Twelve of the C5b9+ patients also had deposition of C4d in GC and PTC. C4d deposits along GC and PTC were not associated with death-censored allograft survival (p = 0.42 and 0.69, respectively). However, death-censored allograft survival was significantly lower in patients with global and diffuse deposition of C5b9 in GC than those with a segmental pattern or no deposition (median survival after ABMR diagnosis, 6 months, 40.5 months and 44 months, respectively; p = 0.015). Double contour of glomerular basement membrane was diagnosed earlier after transplantation in C5b9+ ABMR than in C5b9– ABMR (median time after transplantation, 28 vs. 85 months; p = 0.058). In conclusion, we identified a new pattern of C5b9+ ABMR, associated with early onset of glomerular basement membrane duplication and poor allograft survival. Complement inhibitors might be a therapeutic option for this subgroup of patients

    Daprodustat for the treatment of anemia in patients not undergoing dialysis

    Get PDF
    Background: Daprodustat is an oral hypoxia-inducible factor prolyl hydroxylase inhibitor. In patients with chronic kidney disease (CKD) who are not undergoing dialysis, the efficacy and safety of daprodustat, as compared with the conventional erythropoiesis-stimulating agent darbepoetin alfa, are unknown. Methods: In this randomized, open-label, phase 3 trial with blinded adjudication of cardiovascular outcomes, we compared daprodustat with darbepoetin alfa for the treatment of anemia in patients with CKD who were not undergoing dialysis. The primary outcomes were the mean change in the hemoglobin level from baseline to weeks 28 through 52 and the first occurrence of a major adverse cardiovascular event (MACE; a composite of death from any cause, nonfatal myocardial infarction, or nonfatal stroke). Results: Overall, 3872 patients were randomly assigned to receive daprodustat or darbepoetin alfa. The mean (±SD) baseline hemoglobin levels were similar in the two groups. The mean (±SE) change in the hemoglobin level from baseline to weeks 28 through 52 was 0.74±0.02 g per deciliter in the daprodustat group and 0.66±0.02 g per deciliter in the darbepoetin alfa group (difference, 0.08 g per deciliter; 95% confidence interval [CI], 0.03 to 0.13), which met the prespecified noninferiority margin of −0.75 g per deciliter. During a median follow-up of 1.9 years, a first MACE occurred in 378 of 1937 patients (19.5%) in the daprodustat group and in 371 of 1935 patients (19.2%) in the darbepoetin alfa group (hazard ratio, 1.03; 95% CI, 0.89 to 1.19), which met the prespecified noninferiority margin of 1.25. The percentages of patients with adverse events were similar in the two groups. Conclusions: Among patients with CKD and anemia who were not undergoing dialysis, daprodustat was noninferior to darbepoetin alfa with respect to the change in the hemoglobin level from baseline and with respect to cardiovascular outcomes. (Funded by GlaxoSmithKline; ASCEND-ND ClinicalTrials.gov number, NCT02876835

    Race-free estimated glomerular filtration rate equation in kidney transplant recipients:development and validation study

    Get PDF
    OBJECTIVE: To compare the performance of a newly developed race-free kidney recipient specific glomerular filtration rate (GFR) equation with the three current main equations for measuring GFR in kidney transplant recipients.DESIGN: Development and validation study SETTING: 17 cohorts in Europe, the United States, and Australia (14 transplant centres, three clinical trials).PARTICIPANTS: 15 489 adults (3622 in development cohort (Necker, Saint Louis, and Toulouse hospitals, France), 11 867 in multiple external validation cohorts) who received kidney transplants between 1 January 2000 and 1 January 2021.MAIN OUTCOME MEASURE: The main outcome measure was GFR, measured according to local practice. Performance of the GFR equations was assessed using P 30 (proportion of estimated GFR (eGFR) within 30% of measured GFR (mGFR)) and correct classification (agreement between eGFR and mGFR according to GFR stages). The race-free equation, based on creatinine level, age, and sex, was developed using additive and multiplicative linear regressions, and its performance was compared with the three current main GFR equations: Modification of Diet in Renal Disease (MDRD) equation, Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) 2009 equation, and race-free CKD-EPI 2021 equation. RESULTS: The study included 15 489 participants, with 50 464 mGFR and eGFR values. The mean GFR was 53.18 mL/min/1.73m 2 (SD 17.23) in the development cohort and 55.90 mL/min/1.73m 2 (19.69) in the external validation cohorts. Among the current GFR equations, the race-free CKD-EPI 2021 equation showed the lowest performance compared with the MDRD and CKD-EPI 2009 equations. When race was included in the kidney recipient specific GFR equation, performance did not increase. The race-free kidney recipient specific GFR equation showed significantly improved performance compared with the race-free CKD-EPI 2021 equation and performed well in the external validation cohorts (P 30 ranging from 73.0% to 91.3%). The race-free kidney recipient specific GFR equation performed well in several subpopulations of kidney transplant recipients stratified by race (P 30 73.0-91.3%), sex (72.7-91.4%), age (70.3-92.0%), body mass index (64.5-100%), donor type (58.5-92.9%), donor age (68.3-94.3%), treatment (78.5-85.2%), creatinine level (72.8-91.3%), GFR measurement method (73.0-91.3%), and timing of GFR measurement post-transplant (72.9-95.5%). An online application was developed that estimates GFR based on recipient's creatinine level, age, and sex (https://transplant-prediction-system.shinyapps.io/eGFR_equation_KTX/). CONCLUSION: A new race-free kidney recipient specific GFR equation was developed and validated using multiple, large, international cohorts of kidney transplant recipients. The equation showed high accuracy and outperformed the race-free CKD-EPI 2021 equation that was developed in individuals with native kidneys.TRIAL REGISTRATION: ClinicalTrials.gov NCT05229939.</p

    Both Monoclonal and Polyclonal Immunoglobulin Contingents Mediate Complement Activation in Monoclonal Gammopathy Associated-C3 Glomerulopathy

    Get PDF
    C3 glomerulopathy (C3G) results from acquired or genetic abnormalities in the complement alternative pathway (AP). C3G with monoclonal immunoglobulin (MIg-C3G) was recently included in the spectrum of “monoclonal gammopathy of renal significance.” However, mechanisms of complement dysregulation in MIg-C3G are not described and the pathogenic effect of the monoclonal immunoglobulin is not understood. The purpose of this study was to investigate the mechanisms of complement dysregulation in a cohort of 41 patients with MIg-C3G. Low C3 level and elevated sC5b-9, both biomarkers of C3 and C5 convertase activation, were present in 44 and 78% of patients, respectively. Rare pathogenic variants were identified in 2/28 (7%) tested patients suggesting that the disease is acquired in a large majority of patients. Anti-complement auto-antibodies were found in 20/41 (49%) patients, including anti-FH (17%), anti-CR1 (27%), anti-FI (5%) auto-antibodies, and C3 Nephritic Factor (7%) and were polyclonal in 77% of patients. Using cofactor assay, the regulation of the AP was altered in presence of purified IgG from 3/9 and 4/7 patients with anti-FH or anti-CR1 antibodies respectively. By using fluid and solid phase AP activation, we showed that total purified IgG of 22/34 (65%) MIg-C3G patients were able to enhance C3 convertase activity. In five documented cases, we showed that the C3 convertase enhancement was mostly due to the monoclonal immunoglobulin, thus paving the way for a new mechanism of complement dysregulation in C3G. All together the results highlight the contribution of both polyclonal and monoclonal Ig in MIg-C3G. They provide direct insights to treatment approaches and opened up a potential way to a personalized therapeutic strategy based on chemotherapy adapted to the B cell clone or immunosuppressive therapy

    Predictive factors for recurrence of atypical hemolytic uremic syndrome and C3 glomerulonephritis after renal transplantation

    No full text
    Le syndrome hĂ©molytique et urĂ©mique atypique (SHU) et les glomĂ©rulonĂ©phrites Ă  dĂ©pĂŽts de C3 sont des pathologies rares. Le SHU atypique est une pathologie aigĂŒe sĂ©vĂšre, responsable de microangiopathie thrombotique rĂ©nale glomĂ©rulaire et artĂ©riolaire. La glomĂ©rulonĂ©phrite Ă  dĂ©pĂŽts de C3 (GNC3) est une pathologie chronique responsable d’une protĂ©inurie et d’une insuffisance rĂ©nale chronique qui Ă©volue sur plusieurs annĂ©es. Ces pathologies sont secondaires soit Ă  un dĂ©faut de rĂ©gulation soit Ă  une hyperactivation de la voie alterne du complĂ©ment. Dans ces deux pathologies, le risque de rĂ©cidive aprĂšs transplantation rĂ©nale est Ă©levĂ© et responsable d’une perte de greffon. L’objectif de ma thĂšse a Ă©tĂ© de rechercher des marqueurs prĂ©dictifs de rĂ©cidive et de sĂ©vĂ©ritĂ© conditionnant le pronostic de la transplantation rĂ©nale pour ces deux pathologies complĂ©ment–dĂ©pendantes. Une approche Ă  la fois clinique, biologique et gĂ©nĂ©tique a permis de dĂ©terminer pour le SHU atypique : les facteurs de risque gĂ©nĂ©tique de rĂ©cidive, de dĂ©terminer pour la premiĂšre fois que la prĂ©sence de l’haplotype homozygote du Facteur H (gtgt) est associĂ©e Ă  risque de rĂ©cidive et de prĂ©ciser les consĂ©quences fonctionnelles des mutations. Ce travail a contribuĂ© Ă  Ă©tablir les recommandations du traitement chez les patients transplantĂ©s pour un SHU atypique. Les facteurs de risque de rĂ©cidives des glomĂ©rulonĂ©phrites Ă  C3 ont Ă©tĂ© recherchĂ©s Ă  partir d’une large cohorte de patients que j’ai crĂ©Ă©e. Ce travail a permis de mettre en Ă©vidence une corrĂ©lation entre l’hyperactivation de la voie alterne du complĂ©ment (Ă©lĂ©vation du C5b9 soluble, C3 plasmatique bas), la prĂ©sence de dĂ©pĂŽts (C3, C5b9) dans le rein en post transplantation et la sĂ©vĂ©ritĂ© de la rĂ©cidive. La consommation de C3 en phase fluide, la prĂ©sence d’un sC5b9 Ă©levĂ© et la prĂ©sence de C5b9 in situ sont associĂ©es Ă  une rĂ©cidive sĂ©vĂšre. Enfin, j’ai participĂ© Ă  la premiĂšre description de mutations de DGKE dans le SHU atypique. J’ai mis au point la technique d’immunohistochimie permettant de dĂ©tecter et localiser DGKE dans les cellules endothĂ©liales et podocytaires rĂ©nales. Ce travail a permis de montrer que le DGKE est prĂ©sent dans l’endothĂ©lium rĂ©nal, Ă©lĂ©ment essentiel du mĂ©canisme physiopathologique du SHU atypique. Le DGKE est apportĂ© par le rein transplantĂ© et permet de corriger le risque de rĂ©cidive aprĂšs transplantation chez les patients dĂ©ficitaires.Pas de rĂ©sumĂ© en anglai

    Targeting the complement cascade: novel treatments coming down the pike

    No full text
    International audienceThe complement cascade is a vital component of both the innate and adaptive immune systems. Complement activation also contributes to the pathogenesis of many diseases, however, and the kidney is particularly susceptible to complement-mediated injury. Drugs that block complement activation can rapidly reduce tissue inflammation and also attenuate the adaptive immune response to foreign and tissue antigens. Eculizumab is a monoclonal antibody that prevents the cleavage of C5. It has been approved for the treatment of atypical hemolytic uremic syndrome, and it has been used in selected patients with other kidney diseases. Many additional drugs are also in development for blocking the complement cascade, including new monoclonal antibodies, recombinant proteins, small molecules, and small interfering RNA agents. Validation of these new drugs as effective treatments for kidney diseases faces several challenges. Many complement-mediated kidney diseases are rare, so it is not feasible to test all of the new drugs in numerous different rare diseases. The onset and course of the diseases are heterogeneous; many of these diseases also carry a lifelong risk of recurrence, and it is not clear how long complement inhibition must be maintained. In spite of these challenges, new therapeutic options for targeting the complement system will likely become available in the near future and may prove useful for treating patients with kidney disease

    The Complement System and Antibody-Mediated Transplant Rejection

    No full text
    International audienceComplement activation is an important cause of tissue injury in patients with Ab-mediated rejection (AMR) of transplanted organs. Complement activation triggers a strong inflammatory response, and it also generates tissue-bound and soluble fragments that are clinically useful markers of inflammation. The detection of complement proteins deposited within transplanted tissues has become an indispensible biomarker of AMR, and several assays have recently been developed to measure complement activation by Abs reactive to specific donor HLA expressed within the transplant. Complement inhibitors have entered clinical use and have shown efficacy for the treatment of AMR. New methods of detecting complement activation within transplanted organs will improve our ability to diagnose and monitor AMR, and they will also help guide the use of complement inhibitory drugs
    • 

    corecore