Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
2.
Blood ; 137(18): 2438-2449, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33270832

ABSTRACT

The optimal duration of eculizumab treatment in patients with atypical hemolytic uremic syndrome (aHUS) remains poorly defined. We conducted a prospective national multicenter open-label study to assess eculizumab discontinuation in children and adults with aHUS. Fifty-five patients (including 19 children) discontinued eculizumab (mean treatment duration, 16.5 months). Twenty-eight patients (51%) had rare variants in complement genes, mostly in MCP (n = 12; 22%), CFH (n = 6; 11%), and CFI (n = 6; 10%). At eculizumab discontinuation, 17 (30%) and 4 patients (7%) had stage 3 and 4 chronic kidney disease, respectively. During follow-up, 13 patients (23%; 6 children and 7 adults) experienced aHUS relapse. In multivariable analysis, female sex and presence of a rare variant in a complement gene were associated with an increased risk of aHUS relapse, whereas requirement for dialysis during a previous episode of acute aHUS was not. In addition, increased sC5b-9 plasma level at eculizumab discontinuation was associated with a higher risk of aHUS relapse in all patients and in the subset of carriers with a complement gene rare variant, both by log-rank test and in multivariable analysis. Of the 13 relapsing patients, all of whom restarted eculizumab, 11 regained their baseline renal function and 2 had a worsening of their preexisting chronic kidney disease, including 1 patient who progressed to end-stage renal disease. A strategy of eculizumab discontinuation in aHUS patients based on complement genetics is reasonable and safe. It improves the management and quality of life of a sizeable proportion of aHUS patients while reducing the cost of treatment. This trial was registered at www.clinicaltrials.gov as #NCT02574403.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/drug therapy , Complement Inactivating Agents/therapeutic use , Withholding Treatment/statistics & numerical data , Adolescent , Adult , Atypical Hemolytic Uremic Syndrome/metabolism , Atypical Hemolytic Uremic Syndrome/pathology , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Male , Prognosis , Prospective Studies , Survival Rate , Young Adult
3.
Blood ; 136(19): 2103-2117, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32808006

ABSTRACT

Pregnancy and postpartum are high-risk periods for different forms of thrombotic microangiopathy (TMA). However, the management of pregnancy-associated TMA remains ill defined. This report, by an international multidisciplinary working group of obstetricians, nephrologists, hematologists, intensivists, neonatologists, and complement biologists, summarizes the current knowledge of these potentially severe disorders and proposes a practical clinical approach to diagnose and manage an episode of pregnancy-associated TMA. This approach takes into account the timing of TMA in pregnancy or postpartum, coexisting symptoms, first-line laboratory workup, and probability-based assessment of possible causes of pregnancy-associated TMA. Its aims are: to rule thrombotic thrombocytopenic purpura (TTP) in or out, with urgency, using ADAMTS13 activity testing; to consider alternative disorders with features of TMA (preeclampsia/eclampsia; hemolysis elevated liver enzymes low platelets syndrome; antiphospholipid syndrome); or, ultimately, to diagnose complement-mediated atypical hemolytic uremic syndrome (aHUS; a diagnosis of exclusion). Although they are rare, diagnosing TTP and aHUS associated with pregnancy, and postpartum, is paramount as both require urgent specific treatment.


Subject(s)
ADAMTS13 Protein/metabolism , Pregnancy Complications/physiopathology , Thrombotic Microangiopathies/diagnosis , Thrombotic Microangiopathies/therapy , Disease Management , Female , Humans , International Agencies , Pregnancy , Research Report , Thrombotic Microangiopathies/etiology , Thrombotic Microangiopathies/metabolism
4.
J Am Soc Nephrol ; 30(12): 2449-2463, 2019 12.
Article in English | MEDLINE | ID: mdl-31575699

ABSTRACT

BACKGROUND: Atypical hemolytic uremic syndrome (HUS) is associated with high recurrence rates after kidney transplant, with devastating outcomes. In late 2011, experts in France recommended the use of highly individualized complement blockade-based prophylaxis with eculizumab to prevent post-transplant atypical HUS recurrence throughout the country. METHODS: To evaluate this strategy's effect on kidney transplant prognosis, we conducted a retrospective multicenter study from a large French nationwide registry, enrolling all adult patients with atypical HUS who had undergone complement analysis and a kidney transplant since January 1, 2007. To assess how atypical HUS epidemiology in France in the eculizumab era evolved, we undertook a population-based cohort study that included all adult patients with atypical HUS (n=397) between 2007 and 2016. RESULTS: The first study included 126 kidney transplants performed in 116 patients, 58.7% and 34.1% of which were considered to be at a high and moderate risk of atypical HUS recurrence, respectively. Eculizumab prophylaxis was used in 52 kidney transplants, including 39 at high risk of recurrence. Atypical HUS recurred after 43 (34.1%) of the transplants; in four cases, patients had received eculizumab prophylaxis and in 39 cases they did not. Use of prophylactic eculizumab was independently associated with a significantly reduced risk of recurrence and with significantly longer graft survival. In the second, population-based cohort study, the proportion of transplant recipients among patients with ESKD and atypical HUS sharply increased between 2012 and 2016, from 46.2% to 72.3%, and showed a close correlation with increasing eculizumab use among the transplant recipients. CONCLUSIONS: Results from this observational study are consistent with benefit from eculizumab prophylaxis based on pretransplant risk stratification and support the need for a rigorous randomized trial.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/drug therapy , Complement Inactivating Agents/therapeutic use , Kidney Transplantation , Adult , Atypical Hemolytic Uremic Syndrome/epidemiology , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/surgery , Complement C3b Inactivator Proteins/genetics , Complement System Proteins/analysis , Female , France , Graft Survival/drug effects , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Mutant Chimeric Proteins/genetics , Preoperative Care , Proportional Hazards Models , Recurrence , Registries , Retrospective Studies , Secondary Prevention
5.
Euro Surveill ; 24(8)2019 Feb.
Article in English | MEDLINE | ID: mdl-30808442

ABSTRACT

IntroductionHaemolytic uraemic syndrome (HUS) related to Shiga toxin-producing Escherichia coli (STEC) is the leading cause of acute renal failure in young children. In France, HUS surveillance in children aged < 15 years was implemented starting from 1996.AimWe present the results of this surveillance between 2007 and 2016.MethodsA voluntary nationwide network of 32 paediatric departments notifies cases. Two national reference centres perform microbiological STEC confirmation.ResultsOver the study period, the paediatric HUS incidence rate (IR) was 1.0 per 100,000 children-years, with a median of 116 cases/year. In 2011, IR peaked at 1.3 per 100,000 children-years, and decreased to 1.0 per 100,000 children-years in 2016. STEC O157 associated HUS peaked at 37 cases in 2011 and decreased to seven cases in 2016. Cases of STEC O26-associated HUS have increased since 2010 and STEC O80 associated HUS has emerged since 2012, with 28 and 18 cases respectively reported in 2016. Four STEC-HUS food-borne outbreaks were detected (three STEC O157 linked to ground beef and raw-milk cheese and one STEC O104 linked to fenugreek sprouts). In addition, two outbreaks related to person-to-person transmission occurred in distinct kindergartens (STEC O111 and O26).ConclusionsNo major changes in HUS IRs were observed over the study period of 10 years. However, changes in the STEC serogroups over time and the outbreaks detected argue for continuing epidemiological and microbiological surveillance.


Subject(s)
Disease Outbreaks , Escherichia coli Infections/microbiology , Feces/microbiology , Hemolytic-Uremic Syndrome/epidemiology , Hemolytic-Uremic Syndrome/microbiology , Shiga-Toxigenic Escherichia coli/isolation & purification , Child , Escherichia coli Infections/diagnosis , Escherichia coli Infections/epidemiology , Escherichia coli Proteins , France/epidemiology , Hemolytic-Uremic Syndrome/complications , Humans , Incidence , Infant , Male , Population Surveillance , Serologic Tests , Sex Distribution , Shiga Toxins
6.
Clin Kidney J ; 12(1): 106-109, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30746136

ABSTRACT

Haemolytic uraemic syndrome (HUS) is a rare complication of invasive infection by Streptococcus pneumoniae (SP-HUS), especially in adults. Here we report an unusual case of a 53-year-old man presenting SP-HUS with severe multivisceral involvement. After failure of supportive care and plasma exchanges, eculizumab (anti-C5 antibody) resulted in a favourable outcome.

7.
Clin J Am Soc Nephrol ; 14(3): 364-377, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30674459

ABSTRACT

BACKGROUND AND OBJECTIVES: Inherited complement hyperactivation is critical for the pathogenesis of atypical hemolytic uremic syndrome (HUS) but undetermined in postdiarrheal HUS. Our aim was to investigate complement activation and variants of complement genes, and their association with disease severity in children with Shiga toxin-associated HUS. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Determination of complement biomarkers levels and next-generation sequencing for the six susceptibility genes for atypical HUS were performed in 108 children with a clinical diagnosis of post-diarrheal HUS (75 Shiga toxin-positive, and 33 Shiga toxin-negative) and 80 French controls. As an independent control cohort, we analyzed the genotypes in 503 European individuals from the 1000 Genomes Project. RESULTS: During the acute phase of HUS, plasma levels of C3 and sC5b-9 were increased, and half of patients had decreased membrane cofactor protein expression, which normalized after 2 weeks. Variants with minor allele frequency <1% were identified in 12 Shiga toxin-positive patients with HUS (12 out of 75, 16%), including pathogenic variants in four (four out of 75, 5%), with no significant differences compared with Shiga toxin-negative patients with HUS and controls. Pathogenic variants with minor allele frequency <0.1% were found in three Shiga toxin-positive patients with HUS (three out of 75, 4%) versus only four European controls (four out of 503, 0.8%) (odds ratio, 5.2; 95% confidence interval, 1.1 to 24; P=0.03). The genetic background did not significantly affect dialysis requirement, neurologic manifestations, and sC5b-9 level during the acute phase, and incident CKD during follow-up. However, the only patient who progressed to ESKD within 3 years carried a factor H pathogenic variant. CONCLUSIONS: Rare variants and complement activation biomarkers were not associated with severity of Shiga toxin-associated HUS. Only pathogenic variants with minor allele frequency <0.1% are more frequent in Shiga toxin-positive patients with HUS than in controls.


Subject(s)
Atypical Hemolytic Uremic Syndrome/genetics , Complement Activation/genetics , Complement System Proteins/genetics , Escherichia coli Infections/genetics , Genetic Variation , Shiga-Toxigenic Escherichia coli/pathogenicity , Age Factors , Atypical Hemolytic Uremic Syndrome/immunology , Atypical Hemolytic Uremic Syndrome/microbiology , Child, Preschool , Disease Progression , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Female , France , Gene Frequency , Genetic Predisposition to Disease , Host-Pathogen Interactions , Humans , Infant , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/immunology , Kidney Failure, Chronic/microbiology , Male , Phenotype , Prognosis , Retrospective Studies , Risk Assessment , Risk Factors , Shiga-Toxigenic Escherichia coli/immunology , Time Factors
8.
Semin Hematol ; 55(3): 150-158, 2018 07.
Article in English | MEDLINE | ID: mdl-30032752

ABSTRACT

The dissection of the pathogenic mechanisms of the various forms of the hemolytic uremic syndrome (HUS) has paved the way for the design of specific efficacious treatments. Such mechanistic approach led to a revolution in the management of atypical HUS with the use of the first-in class C5 blocker, eculizumab. The availability of this anticomplement drug has also raised unsettled questions regarding the cost or burden and optimal duration of therapy and its use in secondary HUS. The efficacy of eculizumab in Shiga toxin producing Escherichia coli-associated HUS is not to date established and the results of ongoing prospective studies are eagerly awaited. Nevertheless, the emergence of anticomplement therapies (eculizumab and other drugs in development) has transformed our approach of HUS.


Subject(s)
Atypical Hemolytic Uremic Syndrome/drug therapy , Complement Inactivator Proteins/therapeutic use , Atypical Hemolytic Uremic Syndrome/pathology , Complement Inactivator Proteins/pharmacology , Humans , Treatment Outcome
9.
Am J Kidney Dis ; 72(1): 84-92, 2018 07.
Article in English | MEDLINE | ID: mdl-29429752

ABSTRACT

BACKGROUND: Cases reports and small series of patients with C3 glomerulopathy have reported variable efficacy of eculizumab. STUDY DESIGN: Case series of C3 glomerulopathy. SETTING & PARTICIPANTS: Pediatric and adult patients with C3 glomerulopathy treated with eculizumab between 2010 and 2016 were identified through the C3 glomerulopathy French registry database, and a questionnaire was sent to participating French pediatric and adult nephrology centers, as well as one pediatric referral center in Québec, Canada. OUTCOMES: Global or partial clinical renal response. MEASUREMENTS: Evolution of serum creatinine and proteinuria values. RESULTS: 26 patients (13 children/adolescents) were included. 22 (85%) patients had received steroids, plasma exchange, or immunosuppressive therapy before eculizumab, and 3 of them had rapid progression of their kidney disease despite treatment. At the initiation of eculizumab therapy, 11 (42%) patients had chronic kidney disease, 7 (27%) had rapidly progressive disease, and 3 (12%) required dialysis. After eculizumab treatment (median duration, 14 months), 6 (23%) patients had a global clinical response; 6 (23%), a partial clinical response; and 14 (54%), no response. Compared with those who had a partial clinical or no response, patients who had a global clinical response had lower estimated glomerular filtration rates, a more rapidly progressive course, and more extracapillary proliferation on kidney biopsy. Age, extent of renal fibrosis, frequency of nephrotic syndrome, low serum C3 and C3 nephritic factor and elevated soluble C5b-9 concentrations, or complement gene variants did not differ between responders and nonresponders. LIMITATIONS: Retrospective design without a control group, relatively small number of cases, inclusion of pediatric and adult cases. CONCLUSIONS: Eculizumab appears to be a potential treatment for patients with crescentic rapidly progressive C3 glomerulopathy. Its benefit in patients with non-rapidly progressing forms seems to be limited.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Complement C3/metabolism , Glomerulonephritis, Membranoproliferative/blood , Glomerulonephritis, Membranoproliferative/drug therapy , Adolescent , Adult , Aged , Child , Female , Follow-Up Studies , Glomerulonephritis, Membranoproliferative/diagnosis , Humans , Male , Middle Aged , Retrospective Studies , Treatment Outcome , Young Adult
10.
Clin J Am Soc Nephrol ; 12(8): 1237-1247, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28596415

ABSTRACT

BACKGROUND: Pregnancy is associated with various forms of thrombotic microangiopathy, including hemolytic uremic syndrome. A previous small French study suggested that pregnancy-associated hemolytic uremic syndrome was to be included in the spectrum of atypical hemolytic uremic syndrome linked to complement alternative pathway dysregulation. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: We sought to retrospectively analyze the presentation, outcome, and frequency of complement alternative pathway gene variants in a larger international (France, United Kingdom, Italy) cohort of patients with pregnancy-associated hemolytic uremic syndrome. RESULTS: Eighty-seven patients with pregnancy-associated hemolytic uremic syndrome were included. Hemolytic uremic syndrome occurred mainly during the first pregnancy (58%) and in the postpartum period (76%). At diagnosis, 56 (71%) patients required dialysis. Fifty-six (78%) patients underwent plasma exchanges, 21 (41%) received plasma infusions, and four (5%) received eculizumab. During follow-up (mean duration of 7.2 years), 41 (53%) patients reached ESRD, 15 (19%) had CKD, and 18 (28%) patients experienced hemolytic uremic syndrome relapse. Twenty-four patients (27%) received a kidney transplant and a recurrence of hemolytic uremic syndrome occurred in 13 (54%) patients. Variants in complement genes were detected in 49 (56%) patients, mainly in the CFH (30%) and CFI genes (9%). CONCLUSIONS: Pregnancy-associated hemolytic uremic syndrome and atypical hemolytic uremic syndrome nonrelated to pregnancy have the same severity at onset and during follow-up and the same frequency of complement gene variants.


Subject(s)
Postpartum Period , Pregnancy Complications , Adolescent , Adult , Antibodies, Monoclonal, Humanized/therapeutic use , Complement Activation/drug effects , Complement Activation/genetics , Complement Factor H/genetics , Complement Factor I/genetics , Complement Inactivating Agents/therapeutic use , Disease Progression , Europe , Female , Genetic Predisposition to Disease , Genetic Variation , Hemolytic-Uremic Syndrome/complications , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/therapy , Humans , Kidney Failure, Chronic/etiology , Middle Aged , Phenotype , Plasma Exchange , Pregnancy , Pregnancy Complications/genetics , Pregnancy Complications/immunology , Pregnancy Complications/therapy , Recurrence , Renal Dialysis , Renal Insufficiency, Chronic/etiology , Retrospective Studies , Time Factors , Treatment Outcome , Young Adult
11.
Lancet ; 390(10095): 681-696, 2017 Aug 12.
Article in English | MEDLINE | ID: mdl-28242109

ABSTRACT

Haemolytic uraemic syndrome is a form of thrombotic microangiopathy affecting predominantly the kidney and characterised by a triad of thrombocytopenia, mechanical haemolytic anaemia, and acute kidney injury. The term encompasses several disorders: shiga toxin-induced and pneumococcus-induced haemolytic uraemic syndrome, haemolytic uraemic syndrome associated with complement dysregulation or mutation of diacylglycerol kinase ɛ, haemolytic uraemic syndrome related to cobalamin C defect, and haemolytic uraemic syndrome secondary to a heterogeneous group of causes (infections, drugs, cancer, and systemic diseases). In the past two decades, experimental, genetic, and clinical studies have helped to decipher the pathophysiology of these various forms of haemolytic uraemic syndrome and undoubtedly improved diagnostic approaches. Moreover, a specific mechanism-based treatment has been made available for patients affected by atypical haemolytic uraemic syndrome due to complement dysregulation. Such treatment is, however, still absent for several other disease types, including shiga toxin-induced haemolytic uraemic syndrome.


Subject(s)
Hemolytic-Uremic Syndrome/therapy , Anti-Bacterial Agents/therapeutic use , Biological Products/therapeutic use , Child , Female , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/etiology , Humans , Kidney Transplantation , Pregnancy , Pregnancy Complications/diagnosis , Pregnancy Complications/etiology , Pregnancy Complications/therapy , Recurrence , Treatment Outcome
12.
Clin J Am Soc Nephrol ; 12(1): 50-59, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27799617

ABSTRACT

BACKGROUND AND OBJECTIVES: The complement inhibitor eculizumab has dramatically improved the outcome of atypical hemolytic uremic syndrome. However, the optimal duration of eculizumab treatment in atypical hemolytic uremic syndrome remains debated. We report on the French atypical hemolytic uremic syndrome working group's first 2-year experience with eculizumab discontinuation in patients with atypical hemolytic uremic syndrome. DESIGN, SETTING, PARTICIPANTS & MEASUREMENTS: Using the French atypical hemolytic uremic syndrome registry database, we retrospectively identified all dialysis-free patients with atypical hemolytic uremic syndrome who discontinued eculizumab between 2010 and 2014 and reviewed their relevant clinical and biologic data. The decision to discontinue eculizumab was made by the clinician in charge of the patient. All patients were closely monitored by regular urine dipsticks and blood tests. Eculizumab was rapidly (24-48 hours) restarted in case of relapse. RESULTS: Among 108 patients treated with eculizumab, 38 patients (nine children and 29 adults) discontinued eculizumab (median treatment duration of 17.5 months). Twenty-one patients (55%) carried novel or rare complement genes variants. Renal recovery under eculizumab was equally good in patients with and those without complement gene variants detected. After a median follow-up of 22 months, 12 patients (31%) experienced atypical hemolytic uremic syndrome relapse. Eight of 11 patients (72%) with complement factor H variants, four of eight patients (50%) with membrane cofactor protein variants, and zero of 16 patients with no rare variant detected relapsed. In relapsing patients, early reintroduction (≤48 hours) of eculizumab led to rapid (<7 days) hematologic remission and a return of serum creatinine to baseline level in a median time of 26 days. At last follow-up, renal function remained unchanged in nonrelapsing and relapsing patients compared with baseline values before eculizumab discontinuation. CONCLUSIONS: Pathogenic variants in complement genes were associated with higher risk of atypical hemolytic uremic syndrome relapse after eculizumab discontinuation. Prospective studies are needed to identify biomarkers predictive of relapse and determine the best strategy of retreatment in relapsing patients.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/drug therapy , Atypical Hemolytic Uremic Syndrome/genetics , Complement System Proteins/genetics , Adolescent , Adult , Aged , Atypical Hemolytic Uremic Syndrome/physiopathology , Child , Child, Preschool , Complement Factor H/genetics , Female , Follow-Up Studies , Humans , Male , Membrane Cofactor Protein/genetics , Middle Aged , Recurrence , Withholding Treatment , Young Adult
13.
Lancet Haematol ; 3(11): e537-e546, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27720178

ABSTRACT

BACKGROUND: Thrombotic thrombocytopenic purpura is a rare thrombotic microangiopathy, related to a severe ADAMTS13 deficiency (a disintegrin and metalloprotease with thromboSpondin type 1 repeats, member 13; activity <10% of normal). Childhood-onset thrombotic thrombocytopenic purpura is very rare and initially often misdiagnosed, especially when ADAMTS13 deficiency is acquired (ie, not linked to inherited mutations of the ADAMTS13 gene). We aimed to investigate initial presentation, management, and outcome of acquired thrombotic thrombocytopenic purpura in children. METHODS: Between Jan 1, 2000, and Dec 31, 2015, we studied a cohort of patients with child-onset and adolescent-onset acquired thrombotic thrombocytopenic purpura included in the French national registry for thrombotic microangiopathies at presentation and during follow up. The inclusion criteria were: first episode before age 18 years; ADAMTS13 activity less than 10% of normal at presentation; positive anti-ADAMTS13 autoantibodies during an episode, or a recovery of ADAMTS13 activity in remission, or both. ADAMTS13 activity and anti-ADAMTS13 autoantibodies were investigated by a central laboratory, and medical records were extensively reviewed to collect clinical and biological features with a standardised form. This study is registered with ClinicalTrials.gov, number NCT00426686. FINDINGS: We enrolled 973 patients with childhood-onset thrombotic microangiopathy, of whom 74 had a severe ADAMTS13 deficiency (activity <10%) at presentation. 24 patients had an inherited thrombotic thrombocytopenic purpura also known as Upshaw-Schulman syndrome and five did not have follow-up data available, thus 45 children had acquired thrombotic thrombocytopenic purpura and were included in our database at presentation. 25 (56%) patients had idiopathic disease and 20 (44%) had miscellaneous associated clinical conditions. At diagnosis, median age was 13 years (IQR 7-16, range 4 months-17 years), with a sex ratio of 2·5 girls to 1 boy. Anti-ADAMTS13 autoantibodies were positive in 37 (82%) of 45 patients (24 [96%] of 25 idiopathic cases and 13 [65%] of 20 non-idiopathic cases). 39 (87%) of 45 patients were given plasma therapy and 21 (47%) received additional rituximab. Four (9%) children died after the first thrombotic thrombocytopenic purpura episode. Long-term follow up of the 41 survivors showed that ten (24%) patients relapsed and systemic lupus erythematosus occurred in two (5%) patients. Preemptive rituximab was used in seven (17%) of 41 patients with acquired thrombotic thrombocytopenic purpura. INTERPRETATION: Our study shows that child-onset and adolescent-onset acquired thrombotic thrombocytopenic purpura have specific clinical, biological and therapeutic features. Long-term follow-up is crucial to prevent relapses of the disease, to identify the occurrence of autoimmune disorders, and to evaluate consequences on social life. Child-onset and adolescent-onset acquired thrombotic thrombocytopenic purpura is a crucial diagnosis in the field of paediatric haematologic cytopenias because it is a life-threatening disease requiring a specific management. FUNDING: Assistance Publique-Hôpitaux de Paris, France.


Subject(s)
ADAMTS13 Protein/deficiency , Purpura, Thrombotic Thrombocytopenic , ADAMTS13 Protein/blood , ADAMTS13 Protein/genetics , Adolescent , Age Factors , Age of Onset , Child , Child, Preschool , Cohort Studies , Female , France , Genetic Predisposition to Disease , Humans , Infant , Male , Purpura, Thrombotic Thrombocytopenic/blood , Purpura, Thrombotic Thrombocytopenic/diagnosis , Purpura, Thrombotic Thrombocytopenic/genetics , Purpura, Thrombotic Thrombocytopenic/therapy , Registries , Rituximab/therapeutic use
14.
Kidney Int ; 90(2): 430-439, 2016 08.
Article in English | MEDLINE | ID: mdl-27342959

ABSTRACT

Dent disease classically combines low-molecular-weight proteinuria, hypercalciuria with nephrocalcinosis, and renal failure. Nephrotic range proteinuria, normal calciuria, and hypokalemia have been rarely reported. It is unknown whether the changes in phenotype observed over time are explained by a decrease in glomerular filtration rate (GFR) or whether there is any phenotype-genotype relationship. To answer this we retrospectively analyzed data from 109 male patients with CLCN5 mutations (Dent-1) and 9 patients with mutation of the OCRL gene (Dent-2). In Dent-1 disease, the estimated GFR decreased with age, by 1.0 to 1.6 ml/min per 1.73 m(2)/yr in the absence and presence of nephrocalcinosis, respectively, with no significant difference. Median values of low-molecular-weight proteinuria were in the nephrotic range and remained at the same level even in late renal disease. End-stage renal disease occurred in 12 patients, at a median age of 40 years. Hypercalciuria decreased with glomerular filtration and was absent in 40% of the patients under 30 and 85% of those over the age of 30. Hypophosphatemia did not resolve with age and calcitriol concentrations were in the upper normal range. Kalemia decreased with age, with half of the patients over the age of 18 presenting with hypokalemia. Thus, no phenotype/genotype correlation was observed in this cohort of patients with Dent disease.


Subject(s)
Chloride Channels/genetics , Genetic Diseases, X-Linked/genetics , Kidney Failure, Chronic/epidemiology , Nephrolithiasis/genetics , Phosphoric Monoester Hydrolases/genetics , Adolescent , Adult , Age Factors , Child , Child, Preschool , Genetic Association Studies , Genetic Diseases, X-Linked/blood , Genetic Diseases, X-Linked/complications , Genetic Diseases, X-Linked/urine , Genotype , Glomerular Filtration Rate , Humans , Hypercalciuria/genetics , Hypercalciuria/urine , Hypophosphatemia/blood , Hypophosphatemia/genetics , Kidney Failure, Chronic/etiology , Male , Middle Aged , Mutation , Nephrolithiasis/blood , Nephrolithiasis/complications , Nephrolithiasis/urine , Phenotype , Proteinuria/genetics , Proteinuria/urine , Retrospective Studies , Young Adult
15.
Pediatr Nephrol ; 31(1): 15-39, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25859752

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) emerged during the last decade as a disease largely of complement dysregulation. This advance facilitated the development of novel, rational treatment options targeting terminal complement activation, e.g., using an anti-C5 antibody (eculizumab). We review treatment and patient management issues related to this therapeutic approach. We present consensus clinical practice recommendations generated by HUS International, an international expert group of clinicians and basic scientists with a focused interest in HUS. We aim to address the following questions of high relevance to daily clinical practice: Which complement investigations should be done and when? What is the importance of anti-factor H antibody detection? Who should be treated with eculizumab? Is plasma exchange therapy still needed? When should eculizumab therapy be initiated? How and when should complement blockade be monitored? Can the approved treatment schedule be modified? What approach should be taken to kidney and/or combined liver-kidney transplantation? How should we limit the risk of meningococcal infection under complement blockade therapy? A pressing question today regards the treatment duration. We discuss the need for prospective studies to establish evidence-based criteria for the continuation or cessation of anticomplement therapy in patients with and without identified complement mutations.


Subject(s)
Atypical Hemolytic Uremic Syndrome/therapy , Nephrology/standards , Adolescent , Age Factors , Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/diagnosis , Atypical Hemolytic Uremic Syndrome/epidemiology , Atypical Hemolytic Uremic Syndrome/immunology , Child , Child, Preschool , Combined Modality Therapy , Complement Activation/drug effects , Consensus , Cooperative Behavior , Drug Monitoring , Humans , Immunologic Factors/therapeutic use , Infant , Infant, Newborn , International Cooperation , Kidney Transplantation , Liver Transplantation , Monitoring, Immunologic , Patient Selection , Plasma Exchange , Predictive Value of Tests , Risk Factors , Treatment Outcome
16.
Hum Mutat ; 36(8): 743-52, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25907713

ABSTRACT

Dent disease is a rare X-linked tubulopathy characterized by low molecular weight proteinuria, hypercalciuria, nephrocalcinosis and/or nephrolithiasis, progressive renal failure, and variable manifestations of other proximal tubule dysfunctions. It often progresses over a few decades to chronic renal insufficiency, and therefore molecular characterization is important to allow appropriate genetic counseling. Two genetic subtypes have been described to date: Dent disease 1 is caused by mutations of the CLCN5 gene, coding for the chloride/proton exchanger ClC-5; and Dent disease 2 by mutations of the OCRL gene, coding for the inositol polyphosphate 5-phosphatase OCRL-1. Herein, we review previously reported mutations (n = 192) and their associated phenotype in 377 male patients with Dent disease 1 and describe phenotype and novel (n = 42) and recurrent mutations (n = 24) in a large cohort of 117 Dent disease 1 patients belonging to 90 families. The novel missense and in-frame mutations described were mapped onto a three-dimensional homology model of the ClC-5 protein. This analysis suggests that these mutations affect the dimerization process, helix stability, or transport. The phenotype of our cohort patients supports and extends the phenotype that has been reported in smaller studies.


Subject(s)
Chloride Channels/genetics , Dent Disease/genetics , Mutation , Animals , Chloride Channels/chemistry , Chloride Channels/metabolism , Cohort Studies , Dent Disease/metabolism , Genetic Association Studies , Humans , Male , Mice , Mice, Knockout , Pedigree
17.
Kidney Int ; 87(5): 1061-73, 2015 May.
Article in English | MEDLINE | ID: mdl-25651368

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is a rare, possibly life-threatening disease characterized by platelet activation, hemolysis and thrombotic microangiopathy (TMA) leading to renal and other end-organ damage. We originally conducted two phase 2 studies (26 weeks and 1 year) evaluating eculizumab, a terminal complement inhibitor, in patients with progressing TMA (trial 1) and those with long duration of aHUS and chronic kidney disease (trial 2). The current analysis assessed outcomes after 2 years (median eculizumab exposure 100 and 114 weeks, respectively). At all scheduled time points, eculizumab inhibited terminal complement activity. In trial 1 with 17 patients, the platelet count was significantly improved from baseline, and hematologic normalization was achieved in 13 patients at week 26, and in 15 patients at both 1 and 2 years. The estimated glomerular filtration rate (eGFR) was significantly improved compared with baseline and year 1. In trial 2 with 20 patients, TMA event-free status was achieved by 16 patients at week 26, 17 patients at year 1, and 19 patients at year 2. Criteria for hematologic normalization were met by 18 patients at each time point. Improvement of 15 ml/min per 1.73 m(2) or more in eGFR was achieved by 1 patient at week 26, 3 patients at 1 year, and 8 patients at 2 years. The mean change in eGFR was not significant compared with baseline, week 26, or year 1. Eculizumab was well tolerated, with no new safety concerns or meningococcal infections. Thus, a 2-year analysis found that the earlier clinical benefits achieved by eculizumab treatment of aHUS were maintained at 2 years of follow-up.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Atypical Hemolytic Uremic Syndrome/drug therapy , Adolescent , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Atypical Hemolytic Uremic Syndrome/blood , Female , Glomerular Filtration Rate , Humans , Male , Middle Aged , Quality of Life , Treatment Outcome , Young Adult
18.
Pediatr Nephrol ; 29(10): 1967-78, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24817340

ABSTRACT

BACKGROUND: In 2009, the European Paediatric Study Group for Haemolytic Uraemic Syndrome (HUS) published a clinical practice guideline for the investigation and initial therapy of diarrhea-negative HUS (now more widely referred to as atypical HUS, aHUS). The therapeutic component of the guideline (comprising early, high-volume plasmapheresis) was derived from anecdotal evidence and expert consensus, and the authors committed to auditing outcome. METHODS: Questionnaires were distributed to pediatric nephrologists across Europe, North America, and the Middle East, who were asked to complete one questionnaire per patient episode of aHUS between July 1, 2009 and December 31, 2010. Comprehensive, anonymous demographic and clinical data were collected. RESULTS: Seventy-one children were reported with an episode of aHUS during the audit period. Six cases occurred on a background of influenza A H1N1 infection. Of 71 patients, 59 (83 %) received plasma therapy within the first 33 days, of whom ten received plasma infusion only. Complications of central venous catheters occurred in 16 out of 51 patients with a catheter in-situ (31 %). Median time to enter hematological remission was 11.5 days, and eight of 71 (11 %) patients did not enter hematological remission by day 33. Twelve patients (17 %) remained dialysis dependent at day 33. CONCLUSIONS: This audit provides a snapshot of the early outcome of a group of children with aHUS in the months prior to more widespread use of eculizumab.


Subject(s)
Atypical Hemolytic Uremic Syndrome/therapy , Guideline Adherence/statistics & numerical data , Adolescent , Child , Child, Preschool , Clinical Audit , Female , Humans , Infant , Infant, Newborn , Male , Practice Guidelines as Topic , Surveys and Questionnaires
19.
Kidney Int ; 85(5): 1019-22, 2014 May.
Article in English | MEDLINE | ID: mdl-24786877

ABSTRACT

Atypical hemolytic uremic syndrome (HUS) secondary to anti-factor H autoantibodies has a poor prognosis. The study by Sinha et al. of a large cohort of Indian children makes a substantial contribution to improved management of this form of HUS by showing that standardized titration of anti-factor H autoantibodies is applicable worldwide and that early treatment initiation and guidance of maintenance treatment by autoantibody titer monitoring significantly improve outcomes.


Subject(s)
Autoantibodies/blood , Blood Proteins/immunology , Complement C3b Inactivator Proteins/immunology , Hemolytic-Uremic Syndrome/therapy , Immunosuppressive Agents/therapeutic use , Plasma Exchange , Time-to-Treatment , Female , Humans , Male
20.
Mol Genet Metab ; 111(3): 314-320, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24440466

ABSTRACT

OBJECTIVE: To establish the safety and efficacy of a new gel formulation of cysteamine hydrochloride (CH) eye drops, for the treatment of corneal complications of nephropathic cystinosis. DESIGN: Open label dose response clinical trial. PARTICIPANTS: Eight patients with infantile nephropathic cystinosis including 4 children, 3 adolescents, and 1 adult (mean age at inclusion, 12.1 ± 4.6 years) treated with CH 0.1% eye drops. INTERVENTION: Patients were treated, in both eyes, with the control CH 0.1% eye drop formulation on average 4 times daily for one month and then switched to Cystadrops® at the same dose frequency. Based on clinical ocular findings, the dose regimen was adapted at D30 and D90 in order to decrease the frequency of instillation. After D90, this dose frequency was maintained, except in cases of crystal density worsening. Patients had a follow-up visit every 6 months during 48 months. MAIN OUTCOME MEASURES: Safety assessment consisted of adverse event and serious adverse event monitoring and recording at each visit. For the efficacy study, the primary endpoint was the corneal cystine crystal density measured with an in vivo confocal microscopy (IVCM) score. RESULTS: All patients completed the study. During the 4-year study period, neither serious adverse events nor significant adverse events related to the study drug were reported. After switching to Cystadrops®, the IVCM total score decreased from baseline to D90 by a mean of 28.6 ± 17.5% (p<0.001). From D90 to M48, the IVCM total score remained stable and significantly decreased as compared to that at D1 despite a reduced dose regimen from D90. At M48, the mean IVCM total score was 8.13 ± 4.15, decreased by a mean 29.9 ± 26.29% from D1 (p = 0.001), with a reduced number of instillations compared to that at D1. The IVCM total score and photophobia were significantly correlated (p = 0.04). CONCLUSION: This study provides evidence that Cystadrops® gel is superior to the CH 0.1% formulation in terms of efficacy and has a good safety profile over a long follow-up period.


Subject(s)
Cysteamine/administration & dosage , Cystine/metabolism , Cystinosis/drug therapy , Administration, Topical , Adolescent , Adult , Child , Cornea/pathology , Crystallization , Cystine/chemistry , Cystinosis/metabolism , Cystinosis/pathology , Female , Gels/administration & dosage , Humans , Male , Ophthalmic Solutions/administration & dosage , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...