1
|
Uriol-Rivera MG, Ernst FR, Booth JN, Comas À, Gasteyger C, Tomazos I, Lum C, Wang Y, Ávila AI. PLASMIC score to aid diagnosis of aHUS: an analysis of C5 inhibitor clinical trials and the PINC AI™ healthcare database. BMC Nephrol 2025; 26:241. [PMID: 40375186 PMCID: PMC12080142 DOI: 10.1186/s12882-025-04156-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 04/25/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Atypical hemolytic uremic syndrome (aHUS) is a thrombotic microangiopathy (TMA) that can lead to end organ damage and death without treatment. The ability to rapidly distinguish aHUS from other forms of TMA is key for optimal patient management. The PLASMIC Score was developed to identify individuals with thrombotic thrombocytopenic purpura (TTP), a TMA subtype characterized by severe ADAMTS13 deficiency (< 10%), using 7 commonly available laboratory variables and aspects of the patient's medical history. This study aimed to assess the distribution of PLASMIC Scores in patients with known aHUS, and evaluate the utility of the PLASMIC Score in the diagnostic pathway of aHUS in patients with confirmed TMA. METHODS Data from eculizumab (NCT01194973) and ravulizumab (NCT02949128) clinical trials were utilized to calculate and evaluate PLASMIC Score distribution in aHUS patients. Real-world patient-level data from the PINC AI™ Healthcare Database (PHD) were used to evaluate the performance of the PLASMIC Score in identifying aHUS in patients with documented TMA diagnoses and renal impairment (primary analysis population; n = 110), and subsequent sensitivity analyses were performed in alternative populations. RESULTS A total of 94 aHUS patients from the eculizumab and ravulizumab clinical trials dataset were evaluated; 18/36 (50.0%) and 27/58 (46.6%) patients in the eculizumab and ravulizumab trials, respectively, had a PLASMIC Score of 4, and most patients (~ 85%) had PLASMIC Scores ≤ 5 (range: 3-5), which were distributed similarly between the trials. Among the 110 patients with undifferentiated TMA (primary analysis) from the PHD, a PLASMIC Score cutoff of ≤ 5 yielded sensitivity, specificity and positive predictive value (PPV) and negative predictive values (NPV) of 86.5%, 71.4%, 92.8% and 55.6%, respectively, for identifying probable aHUS. Similar diagnostic performance was observed at a cutoff value of ≤ 5 in further sensitivity analyses. A cutoff value of ≤ 4 yielded a lower PPV (62.9%), yet a higher NPV (85.7%), with only 3 patients misclassified as TTP. CONCLUSION Application of the PLASMIC Score in the aHUS diagnostic pathway may support clinical judgement and ascertain confidence in the earlier identification and subsequent treatment of patients with aHUS, thereby improving patient outcomes.
Collapse
Affiliation(s)
- Miguel G Uriol-Rivera
- Glomerular Diseases Unit, Son Espases University Hospital, Palma de Mallorca, Spain.
| | - Frank R Ernst
- CTI Clinical Trial and Consulting Services, Inc, Covington, KY, USA
| | - John N Booth
- CTI Clinical Trial and Consulting Services, Inc, Covington, KY, USA
| | - Àngels Comas
- Alexion, AstraZeneca Rare Disease, Barcelona, Spain
| | | | | | - Ching Lum
- Alexion, AstraZeneca Rare Disease, Boston, MA, USA
| | - Yan Wang
- Alexion, AstraZeneca Rare Disease, Boston, MA, USA
| | - Ana I Ávila
- Nephrology Department, Dr Peset University Hospital, Valencia, Spain
| |
Collapse
|
2
|
Empitu M, Jayanthi R, Yasin MN, Bouqoufi A. Efficacy and safety of novel complement inhibitors in atypical haemolytic uremic syndrome: a protocol for systematic review and meta-analysis. BMJ Open 2025; 15:e100159. [PMID: 40374233 PMCID: PMC12083363 DOI: 10.1136/bmjopen-2025-100159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/22/2025] [Indexed: 05/17/2025] Open
Abstract
BACKGROUND Atypical Haemolytic Uremic Syndrome (aHUS) is a rare but life-threatening thrombotic microangiopathy. If inadequately managed, aHUS can lead to progressive kidney failure, cardiovascular complications and multiorgan dysfunction, resulting in high healthcare costs and a substantial impact on patients' quality of life. Novel complement inhibitors offer potential advantages, yet comprehensive evidence comparing their efficacy and safety is limited. This protocol elaborates the systematic review plans to evaluate the effectiveness and the drug safety of complement inhibitors in aHUS. METHODS A systematic search will be conducted across PubMed, Embase, Cochrane Library, Web of Science and Scopus to identify relevant studies. Eligible studies include randomised controlled trials (RCTs), observational studies and case series with at least three aHUS patients treated with novel complement inhibitors. Two independent reviewers will perform data extraction and quality assessment using standardised tools, including the Risk of Bias Tool 2 for RCTs and the Newcastle-Ottawa Scale for observational studies. A meta-analysis will be conducted if feasible, utilising a random-effects model to account for study heterogeneity. ETHICS AND DISSEMINATION Ethical approval is not required as only previously published data will be used. Results will be disseminated via peer-reviewed journals and conferences, targeting healthcare professionals and policymakers to support evidence-based decision-making in aHUS management. PROSPERO REGISTRATION NUMBER CRD42025629879.
Collapse
Affiliation(s)
- Maulana Empitu
- Faculty of Medicine, Airlangga University, Surabaya, East Java, Indonesia
- Faculty of Health, Medicine, and Natural Sciences, Airlangga University, Banyuwangi, East Java, Indonesia
| | - Ruthvika Jayanthi
- Faculty of Medicine, Airlangga University, Surabaya, East Java, Indonesia
| | | | - Afaf Bouqoufi
- Ibn Zohr University, Agadir, Souss-Massa-Draa, Morocco
| |
Collapse
|
3
|
Java A, Sparks MA, Kavanagh D. Post-transplant Thrombotic Microangiopathy. J Am Soc Nephrol 2025; 36:940-951. [PMID: 39888686 PMCID: PMC12059091 DOI: 10.1681/asn.0000000645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/28/2025] [Indexed: 02/02/2025] Open
Abstract
Thrombotic microangiopathy (TMA) is a challenging and serious complication of kidney transplantation that significantly affects graft and patient survival, occurring in 0.8%-15% of transplant recipients. TMA is characterized by microangiopathic hemolytic anemia, thrombocytopenia, and organ injury due to endothelial damage and microthrombi formation in small vessels. However, clinical features can range from a renal-limited form, diagnosed only on a kidney biopsy, to full-blown systemic manifestations, which include neurologic, gastrointestinal, and cardiovascular injury. TMA can arise because of genetic or acquired defects such as in complement-mediated TMA or can occur in the context of other conditions like infections, autoimmune diseases, or immunosuppressive drugs, where complement activation may also play a role. Recurrent TMA after kidney transplant is almost always complement-mediated, although complement overactivation may also play a role in de novo post-transplant TMAs associated with ischemia-reperfusion injury, immunosuppressive drugs, antibody-mediated rejection, viral infections, and relapse of autoimmune diseases, such as antiphospholipid antibody syndrome. Differentiating between a complement-mediated process and one triggered by other factors is often challenging but critical to minimize allograft damage because the former is nonresponsive to supportive therapy, needs long-term anticomplement therapy, and has a high risk of recurrence. Given the central role of complement and effect of genetic defects on the risk of recurrence in many forms of post-transplant TMA, genetic testing for complement disorders is key for proper diagnosis and management. Given that complement activation may also play a role in a subset of TMAs associated with other conditions, prompt recognition and timely initiation of anticomplement therapy is equally important. In addition, TMA associated with noncomplement genes, often part of a broader syndromic process with distinct clinical features, has also been described. Early identification and treatment are essential to prevent graft failure and other severe complications. This review explores the pathophysiologic mechanisms underlying various post-transplant TMAs.
Collapse
Affiliation(s)
- Anuja Java
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Matthew A. Sparks
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
- Renal Section, Durham VA Health Care System, Durham, North Carolina
| | - David Kavanagh
- Complement Therapeutics Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- National Renal Complement Therapeutics Centre, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
4
|
Zhong L, Tang S, Pu Z, Chen K, Di W, Hou Y, Yang H. Impact of prophylactic cytomegalovirus immunoglobulin on cytomegalovirus viremia and graft function in ABO-incompatible living donor kidney transplantation: a retrospective analysis. Front Immunol 2025; 16:1562951. [PMID: 40356931 PMCID: PMC12066264 DOI: 10.3389/fimmu.2025.1562951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Background Cytomegalovirus (CMV) infection poses a significant risk to kidney transplant recipients. CMV immunoglobulin shows promising prophylactic effect, particularly in the context of ABO-incompatible transplants. However, its efficacy in preventing CMV viremia remains underexplored. Methods In this retrospective study, we enrolled patients who underwent ABO-incompatible living donor kidney transplantation between May 2021 and September 2023. Prophylactic CMV immunoglobulin was administered at 100 mg/kg weekly for one month in the combined prophylaxis group, while no prophylactic medication was applied in the preemptive therapy group. The primary outcome was measured as the incidence of clinically relevant CMV viremia (CMV DNA >10,000 copies/mL) within one year after transplantation. Both groups received standard preemptive therapy with ganciclovir or valganciclovir after diagnosed with clinically relevant CMV viremia. Results Prophylactic CMV immunoglobulin significantly reduced clinically relevant viremia incidence compared to preemptive therapy group (16.0% vs. 34.0%, P = 0.04). At the end of the follow-up, the combined prophylaxis group showed higher eGFR (56.40 ± 14.19 vs. 47.30 ± 13.01 mL/min/1.73m², P = 0.0014) and lower serum creatinine (146.5 ± 57.07 vs. 171.2 ± 51.48 µmol/L, P = 0.0274). However, no significant differences in renal function were observed between the groups at1,3, or 6 months post-transplantation. Conclusion CMV immunoglobulin represents a promising prophylactic option for reducing clinically relevant CMV viremia incidence and delaying infection onset in ABO-incompatible kidney transplant recipients.
Collapse
Affiliation(s)
- Linhong Zhong
- Department of Hepatobiliary Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Shijie Tang
- Department of Hepatobiliary Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhongping Pu
- Department of Hepatobiliary Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Kai Chen
- Department of Organ Transplantation, Sichuan Provincial Peoples Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenjia Di
- Department of Organ Transplantation, Sichuan Provincial Peoples Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yifu Hou
- Department of Organ Transplantation, Sichuan Provincial Peoples Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hongji Yang
- Department of Hepatobiliary Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Department of Organ Transplantation, Sichuan Provincial Peoples Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
5
|
Yang L, Liu F, Li X, He L, Gu Y, Sun M, Liu Z, Liu Z. Plasma exchange combined with eculizumab in the management of atypical hemolytic uremic in pediatric patients: A case report. Medicine (Baltimore) 2025; 104:e42090. [PMID: 40228287 PMCID: PMC11999395 DOI: 10.1097/md.0000000000042090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/22/2025] [Indexed: 04/16/2025] Open
Abstract
RATIONALE Atypical hemolytic uremic syndrome (aHUS) is a severe rare disease characterized by microvascular hemolytic anemia, thrombocytopenia, and acute renal failure. PATIENT CONCERNS A 10-year-old male presented with symptoms of weakness, jaundice, pallor, swollen eyelids, and no significant lower limb swelling. Laboratory investigations revealed critical hemoglobin levels, fragmented red blood cells, thrombocytopenia, and acute kidney injury. DIAGNOSES The patient was diagnosed with aHUS based on the clinical trial of microvascular hemolytic anemia, thrombocytopenia, and organ damage due to thrombosis, as well as laboratory findings. INTERVENTIONS The patient was treated with a combination of plasma exchange and the drug Eculizumab. Comprehensive nursing care was provided, including the establishment and maintenance of blood purification pipelines, management of complications (such as coagulation, allergic reactions, hypotension, and bleeding), and psychological support for the patient and family. OUTCOMES The patient successfully completed a 3-month treatment regimen with Eculizumab and has been followed up for an additional 10 months without any recurrence of the condition, demonstrating favorable treatment outcomes. LESSONS The novel approach of combining plasma exchange with Eculizumab in pediatric aHUS presents significant nursing challenges, but the case demonstrates the potential benefits of this combination therapy, particularly in the pediatric population in China where Eculizumab has recently become available. Comprehensive nursing care, including managing complications and providing psychological support, is crucial to the successful treatment of aHUS.
Collapse
Affiliation(s)
- Lian Yang
- Department of Nephrology, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Liu
- Wuhan Children’s Hospital West District Comprehensive Service Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaolu Li
- Department of Nephrology, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lanfen He
- Department of Nephrology, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Gu
- Department of Nephrology, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingcan Sun
- Department of Nephrology, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenzhen Liu
- Department of Nephrology, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zeng Liu
- Department of Nephrology, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Yang H, Zhang J, Wu H, Xu H, Xu Z. Treatment of atypical hemolytic uremic syndrome with eculizumab in a patient presenting with neuropsychiatric prodrome: a case report. Front Immunol 2025; 16:1542973. [PMID: 40264782 PMCID: PMC12011752 DOI: 10.3389/fimmu.2025.1542973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/17/2025] [Indexed: 04/24/2025] Open
Abstract
Atypical hemolytic uremic syndrome (aHUS) is a rare thrombotic microangiopathy (TMA) caused by dysregulation of the complement system. It is characterized by microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury. Owing to its diverse and nonspecific clinical manifestations, early diagnosis of the condition is challenging and typically requires excluding other TMA-related conditions, such as thrombotic thrombocytopenic purpura and hemolytic uremic syndrome caused by Escherichia coli infection. Accurate diagnosis relies on the recognition of typical TMA symptoms, laboratory testing, and the exclusion of other conditions. Treatments typically include plasma exchange, supportive care, and complement-targeted therapy. Eculizumab, a complement component 5 inhibitor, plays a crucial role in aHUS treatment in severe cases as well as when traditional interventions fail. In this case report, we described a female Han Chinese patient who developed aHUS following an upper respiratory tract infection, initially presented with intermittent seizures, and received treatment with eculizumab, plasma exchange, and hemodialysis. The patient ultimately remained dialysis-dependent; however, they achieved complete remission for other systemic complications of aHUS. We emphasized in this case report the importance of timely diagnosis and treatment of aHUS as well as the potential value of eculizumab in improving patient outcomes. Furthermore, successful treatment and follow-up results provide insights into the management of this rare disease, including long-term dialysis requirements and disease monitoring after remission. Thus, clinicians can better understand the clinical manifestations of aHUS and its associated diagnostic challenges, treatment strategies, and long-term management needs.
Collapse
Affiliation(s)
| | | | | | - Hongzhao Xu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhonggao Xu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
7
|
Lax N, Davidovits M, Chodick G, Bernfeld Y, Peled O. Eculizumab is efficacious and safe in pediatric patients with various forms of hemolytic uremic syndrome: a retrospective clinical experience of a tertiary center. Front Pharmacol 2025; 16:1535407. [PMID: 40255570 PMCID: PMC12006164 DOI: 10.3389/fphar.2025.1535407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/25/2025] [Indexed: 04/22/2025] Open
Abstract
Background Eculizumab, a terminal complement inhibitor, prevents thrombotic microangiopathy (TMA) and multiorgan damage in hemolytic uremic syndrome (HUS). We evaluated its efficacy and safety in pediatric patients with TMA sub-types: atypical HUS (aHUS), Shiga toxin-producing Escherichia coli (STEC)-HUS, and transplant-associated TMA (TA-TMA). Methods This retrospective study included all pediatric patients treated with eculizumab for HUS at Schneider Children's Medical Center (2011-2020), including those with pre-existing end-stage kidney disease. Clinical and laboratory parameters were analyzed over 28 weeks. The primary endpoint was achievement of complete TMA response, defined by sustained normalization of hematologic parameters and renal function. Secondary endpoints included TMA event-free status and additional clinical improvements. Results Twenty-four pediatric patients (median age 5.8 years) were included: 13 with aHUS, 5 with STEC-HUS, and 6 with TA-TMA. A complete TMA response was achieved in 12 (50%) of the patients overall: 7 (54%) with aHUS, 3 (60%) with STEC-HUS, and 2 (33%) with TA-TMA. TMA event-free status was reached in 15 (63%) patients. Significant improvements were observed in platelet count (63%), lactate dehydrogenase levels (76% within the first week), hemoglobin (60%), and estimated glomerular filtration rate (79%); while CH-50 levels decreased. No severe adverse events were attributed to eculizumab. Chronic kidney disease stage improved for 17 (90%). Conclusion The efficacy and safety of eculizumab for three TMA subtypes in pediatric patients potentially expands its therapeutic applications. The complete TMA response rate in aHUS supports eculizumab as a first-line use, while the response rate in STEC-HUS suggests potential efficacy beyond eculizumab's primary indication. The early hematologic responses and reduced CH-50 levels confirm the role of eculizumab complement-mediated HUS and underscore the need for further research in TA-TMA.
Collapse
Affiliation(s)
- Naama Lax
- Department of Pharmacy, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
| | - Miriam Davidovits
- Institute of Nephrology, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
- Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gabriel Chodick
- Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Maccabi Institute for Research and Innovation, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Yael Bernfeld
- Department of Pharmacy, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
| | - Orit Peled
- Department of Pharmacy, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
- Adelson School of Medicine, Ariel University, Ariel, Israel
| |
Collapse
|
8
|
Vujović A, Sellier-Leclerc AL, Mancuso MC, Boyer O, Awan A, Gargiulo A, Loos S, Fila M, Jankauskiene A, Ariceta G, Kanzelmeyer N, Vidal E, Van Dyck M, Levart TK, Šimánková N, Decramer S, Hofstetter J, Vivarelli M, Sciascia S, van de Kar NC, Schaefer F. Real-world use of complement inhibitors for haemolytic uraemic syndrome: an analysis of the European Rare Kidney Disease Registry cohort. EClinicalMedicine 2025; 82:103159. [PMID: 40224677 PMCID: PMC11987679 DOI: 10.1016/j.eclinm.2025.103159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 04/15/2025] Open
Abstract
Background Although terminal complement inhibitors transformed the prognosis of atypical haemolytic uraemic syndrome (aHUS) from dismal to favourable, treatment approaches vary due to the intermittent disease nature and high costs. Occasionally, complement inhibition is applied in infectious (i)HUS. We aimed to examine real-world C5 inhibitor use and its impact on patient outcomes. Methods This retrospective cohort study used longitudinal data from the European Rare Kidney Disease Registry, collected from 76 nephrology centres across 24 European countries between January 1, 2019 and January 31, 2024. Eligible patients had aHUS or iHUS with onset after January 1, 2011, and/or documented C5 inhibitor use. Exclusions included complement-unrelated HUS, post-transplant HUS, and prophylactic C5 inhibitor use around kidney transplantation. Data, derived from medical records and focused queries, were used to assess C5 inhibitor duration, via time-to-event analysis, and kidney function based on annual creatinine levels. Findings A total of 238 aHUS and 472 patients with iHUS were included in the analysis. C5 inhibition was applied in 76.5% of aHUS and 18.4% of iHUS, with major utilisation differences between countries (p < 0.0001) and less common use in female patients with aHUS (p = 0.0022). Median (interquartile range) treatment duration was 16.1 (3.6-41.2) months in aHUS and 9 (7-32) days in iHUS. After five years, 56% of genetic, 28% of anti-complement factor H (anti-CFH) antibody-mediated, and 23% of aHUS cases with no identified cause remained on treatment. The long-term (>7 years) risk of treatment resumption was 35% in genetic, 15% in aHUS of no identified cause, and 0% in anti-CFH antibody-mediated aHUS. Post-withdrawal aHUS relapses were mostly mild and did not lead to permanent kidney function impairment, ultimately leading to long-term treatment withdrawal in 92.5% of discontinued cases. Interpretation Currently, C5 inhibitors are administered in three-quarters of newly diagnosed patients with aHUS in Europe, with varied utilisation and discontinuation practices. Treatment withdrawal is common and safe, although relapses may occur, particularly in genetic aHUS. However, baseline disease severity, selective use in expert centres, and indication bias affect outcome comparability. Findings must be considered in the context of patient-specific factors and disease severity at the time of treatment decisions. Funding This research was supported by the European Reference Network for Rare Kidney Diseases, funded by the European Union within the framework of the "EU4Health Programme 2021-2027".
Collapse
Affiliation(s)
- Aleksandra Vujović
- Division of Paediatric Nephrology, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Anne-Laure Sellier-Leclerc
- Service de Néphrologie Pédiatriques, Centre de Référence Des Maladies Rénales Rares Néphrogones Filières Maladies Rares ORKID et ERKNet, Hospices Civils de Lyon, Bron, Lyon, France
| | - Maria Cristina Mancuso
- Division of Paediatric Nephrology, Dialysis and Transplantation, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Olivia Boyer
- Paediatric Nephrology, Necker Enfants Malades Hospital, MARHEA Reference Centre, Imagine Institute, Université Paris Cité, France
| | - Atif Awan
- Division of Paediatric Nephrology, Children's Health Ireland at Temple Street, Temple Street, Ireland
| | - Antonio Gargiulo
- Division of Paediatric Nephrology and Dialysis, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sebastian Loos
- Division of Paediatric Nephrology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Fila
- Division of Paediatric Nephrology and Dialysis, CHU Arnaud de Villeneuve, Centre De Référence Des Maladies Rénales Rares du Sud-Ouest (SORARE), Montpellier, France
| | - Augustina Jankauskiene
- Paediatric Centre, Institute of Clinical Medicine, Vilnius University, Vilnius, Lithuania
| | - Gema Ariceta
- Paediatric Nephrology, Vall d'Hebron Hospital, Autonoma University of Barcelona, Barcelona, Spain
| | - Nele Kanzelmeyer
- Department of Paediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Children's Hospital, Hannover, Germany
| | - Enrico Vidal
- Division of Paediatric Nephrology, Department of Women's and Child's Health, University Hospital of Padova, Padua, Italy
| | - Maria Van Dyck
- Division of Paediatric Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Tanja Kersnik Levart
- Division of Paediatric Nephrology, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Naděžda Šimánková
- Department of Paediatrics, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Stephane Decramer
- Department of Paediatric Internal Medicine, Rheumatology and Nephrology, Toulouse University Hospital, Toulouse, France
- Centre De Référence Des Maladies Rénales Rares du Sud-Ouest (SORARE), Toulouse University Hospital, Toulouse, France
- National Institute of Health and Medical Research (INSERM), UMR 1297, Institute of Cardiovascular and Metabolic Disease, Toulouse, France
| | - Jonas Hofstetter
- Division of Paediatric Nephrology, Department of Paediatrics, University of Heidelberg, Heidelberg, Germany
| | - Marina Vivarelli
- Division of Paediatric Nephrology and Dialysis, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Savino Sciascia
- Division of Nephrology, University of Torino-Ospedale HUB Torino Nord, Turin, Italy
| | - Nicole C.A.J. van de Kar
- Division of Paediatric Nephrology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - Franz Schaefer
- Division of Paediatric Nephrology, Department of Paediatrics, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
9
|
Cao W, Liu Y, Zhang XF, Zheng XL. A mutant complement factor H (W1183R) enhances proteolytic cleavage of von Willebrand factor by ADAMTS-13 under shear. J Thromb Haemost 2025; 23:1229-1240. [PMID: 39798927 DOI: 10.1016/j.jtha.2024.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/31/2024] [Accepted: 11/22/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND A loss-of-functional mutation (W1183R) in human complement factor H (CFH) is associated with complement-associated hemolytic uremic syndrome; mice carrying a similar mutation (W1206R) in CFH also develop thrombotic microangiopathy but its plasma von Willebrand factor (VWF) multimer sizes were dramatically reduced. The mechanism underlying such a dramatic change in plasma VWF multimer distribution in these mice is not fully understood. OBJECTIVES To determine the VWF and CFH interaction and how CFH proteins affect VWF multimer distribution. METHODS We employed recombinant protein expression, purification, and various biochemical and biophysical tools. RESULTS Purified recombinant W1183R-CFH but not wild-type (WT) CFH protein enhanced the proteolytic cleavage of both peptidyl and multimeric VWF substrates by recombinant ADAMTS-13 in a concentration-dependent manner. Microscale thermophoresis assay demonstrated that both W1183R-CFH and WT-CFH proteins bound various VWF fragments (eg, AIM-A1, A1-A2-A3, D'D3, D'D3-A1, and D'D3-A1-A2) with high affinities. Optical tweezer experiments further showed a concentration-dependent alteration in the contour length (Lc) and the persistent length (Lp) following pulling VWF-A2 domain in the presence of W1183R-CFH or WT-CFH protein. AlphaFold experiments revealed conformational changes in the VWF-A2, particularly the central region where the cleavage bond resides following addition of W1183R-CFH or WT-CFH protein. CONCLUSION These results demonstrate for the first time that W1183R-CFH but not WT-CFH protein enhances the proteolytic cleavage of VWF by ADAMTS-13 under shear. This may be achieved by mechanic-induced conformational changes of the central A2 domain, leading to an enhanced cleavage of Tyr1605-Met1606 bond by ADAMTS-13 under pathophysiological conditions.
Collapse
Affiliation(s)
- Wenjing Cao
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA; Institute of Reproductive Medicine and Developmental Sciences, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yi Liu
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - X Frank Zhang
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA.
| | - X Long Zheng
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA; Institute of Reproductive Medicine and Developmental Sciences, The University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
10
|
Smith-Jackson K, Walsh P, Zelek WM, Hoyler T, Martinic MM, Thompson G, Gibson BG, Connelly C, Pappworth IY, Murphy MJ, Kavanagh D, Marchbank KJ. The membrane attack complex drives thrombotic microangiopathy in complement mediated atypical hemolytic uremic syndrome. Kidney Int 2025; 107:700-713. [PMID: 39848404 DOI: 10.1016/j.kint.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 11/18/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025]
Abstract
Introduction of complement (C) inhibition into clinical practice has revolutionized the treatment of patients with complement-mediated atypical hemolytic syndrome (aHUS). Our C3D1115N mouse model, engineered around a gain of function point mutation in C3, is associated with complement mediated aHUS in man, allowing us to study the clinical disease in a preclinical model. Backcrossing our model onto C7 deficient and C5aR1 deficient mice enabled further determination of the roles of the C5a-C5aR1 axis and C5b-9 (the membrane attack complex) on kidney disease. C7 deficiency completely abolished both clinical and histological evidence of disease. Removing C5aR1 (CD88) attenuated the risk of developing clinical disease, but mice still developed thrombotic microangiopathy. Therapeutic inhibition strengthened our genetic findings showing both anti-C7 therapy and an oral C5aR1 antagonist, when used before evidence of significant kidney injury, prevented mice from succumbing to disease. However, there was ongoing histological disease within mice treated with the C5aR1 antagonist. Our data suggest that both C5aR1 and C7 play a role in the development of the conditions required for thrombotic microangiopathy of the kidney. While disrupting the C5a-C5aR1 axis is beneficial, our genetic and therapeutic studies showed that thrombotic microangiopathy of the kidney can still develop and ultimately our data confirm that the membrane attack complex is required to develop thrombotic microangiopathy of the kidney. Overall, our study shows that in addition to requiring alternative pathway dysregulation, local generation of membrane attack complex within the kidney is also critical to drive disease pathology in complement-mediated aHUS.
Collapse
Affiliation(s)
- Kate Smith-Jackson
- Complement Therapeutics Research Group, Newcastle University Translational and Clinical Research Institute, the Medical School, Newcastle-upon-Tyne, UK; National Renal Complement Therapeutics Centre, the Royal Victoria Infirmary, Newcastle-upon-Tyne, UK.
| | - Patrick Walsh
- Complement Therapeutics Research Group, Newcastle University Translational and Clinical Research Institute, the Medical School, Newcastle-upon-Tyne, UK; National Renal Complement Therapeutics Centre, the Royal Victoria Infirmary, Newcastle-upon-Tyne, UK
| | - Wioleta M Zelek
- UK Dementia Research Institute Cardiff and Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Thomas Hoyler
- Immunology and Oncology Biology, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | - Marianne M Martinic
- Immunology and Oncology Biology, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | - Gemma Thompson
- Complement Therapeutics Research Group, Newcastle University Translational and Clinical Research Institute, the Medical School, Newcastle-upon-Tyne, UK; National Renal Complement Therapeutics Centre, the Royal Victoria Infirmary, Newcastle-upon-Tyne, UK
| | - Beth G Gibson
- Complement Therapeutics Research Group, Newcastle University Translational and Clinical Research Institute, the Medical School, Newcastle-upon-Tyne, UK; National Renal Complement Therapeutics Centre, the Royal Victoria Infirmary, Newcastle-upon-Tyne, UK
| | - Chloe Connelly
- Complement Therapeutics Research Group, Newcastle University Translational and Clinical Research Institute, the Medical School, Newcastle-upon-Tyne, UK; National Renal Complement Therapeutics Centre, the Royal Victoria Infirmary, Newcastle-upon-Tyne, UK
| | - Isabel Y Pappworth
- Complement Therapeutics Research Group, Newcastle University Translational and Clinical Research Institute, the Medical School, Newcastle-upon-Tyne, UK; National Renal Complement Therapeutics Centre, the Royal Victoria Infirmary, Newcastle-upon-Tyne, UK
| | - Mark J Murphy
- Immunology and Oncology Biology, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | - David Kavanagh
- Complement Therapeutics Research Group, Newcastle University Translational and Clinical Research Institute, the Medical School, Newcastle-upon-Tyne, UK; National Renal Complement Therapeutics Centre, the Royal Victoria Infirmary, Newcastle-upon-Tyne, UK
| | - Kevin J Marchbank
- Complement Therapeutics Research Group, Newcastle University Translational and Clinical Research Institute, the Medical School, Newcastle-upon-Tyne, UK; National Renal Complement Therapeutics Centre, the Royal Victoria Infirmary, Newcastle-upon-Tyne, UK
| |
Collapse
|
11
|
Franchini M, Focosi D, Pezzo MP, Mannucci PM. Catastrophic Thrombosis: A Narrative Review. Semin Thromb Hemost 2025; 51:312-321. [PMID: 39151904 DOI: 10.1055/s-0044-1788790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2024]
Abstract
Catastrophic thrombosis is a severe condition characterized by a hypercoagulable tendency, leading to multiple thromboembolic events in different blood vessels, usually within a short timeframe. Several conditions have been associated with the development of catastrophic thrombosis, including the catastrophic antiphospholipid syndrome, thrombotic anti-platelet factor 4 immune disorders, thrombotic microangiopathies, cancers, the hyper-eosinophilic syndrome, pregnancy, infections, trauma, and drugs. Thrombotic storm represents a medical emergency whose management represents a serious challenge for physicians. Besides the prompt start of anticoagulation, a patient's prognosis depends on early recognition and possible treatment of the underlying condition. In this narrative review, we summarize the main characteristics of catastrophic thrombosis, analyzing the various conditions triggering such life-threatening complication. Finally, an algorithm with the diagnostic workup and the initial management of patients with catastrophic thrombosis is presented.
Collapse
Affiliation(s)
- Massimo Franchini
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, Mantova, Italy
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Italy
| | | | - Pier Mannuccio Mannucci
- Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico and University of Milan, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| |
Collapse
|
12
|
Zagożdżon I, Szczepańska M, Rubik J, Zachwieja K, Musielak A, Bratkowska M, Makulska I, Niwińska K, Leszczyńska B, Bieniaś B, Taranta-Janusz K, Adamczyk-Kipigroch H, Żurowska A. Haemolytic uremic syndrome as a cause of chronic kidney disease stage 5 in children is in retreat: results from the Polish Registry of Kidney Replacement Therapy in children (2000-2023). Pediatr Nephrol 2025; 40:1069-1079. [PMID: 39549043 PMCID: PMC11885394 DOI: 10.1007/s00467-024-06584-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND Haemolytic uremic syndrome (HUS) is a life-threatening disease with a historically poor prognosis in children receiving maintenance kidney replacement therapy (KRT). This study aimed to analyse the incidence and outcome of chronic kidney disease stage 5 (CKD5) due to Escherichia coli-HUS (STEC-HUS) and complement-mediated HUS (CM-HUS) in children, compared with controls with non-HUS CKD5 over the last 24 years. METHODS The study included 1488 children undergoing KRT in Poland between 2000 and 2023. Thirty-nine patients with CM-HUS and 18 with STEC-HUS were identified and analysed for incidence, KRT modality and survival. RESULTS The incidence rate of CKD5 was 0.09 cases/million age-related population (marp) for STEC-HUS and 0.23/marp for CM-HUS, while no new cases have been observed in recent years. CKD5 due to CM-HUS developed significantly earlier from initial HUS manifestation than in STEC-HUS (median 0.2 vs. 9.8 years). CM-HUS was associated with younger age at initiation of KRT compared to STEC-HUS and non-HUS controls (median 6.0 years vs. 10.9 and 10.9 years), with higher risk of death (Hazard Ratio 1.92, 95% confidence interval 0.9-4.13) and worse 5-year kidney graft survival at 77%, 93% and 90%, respectively (p < 0.001). CONCLUSIONS In recent years, both CM-HUS and STEC-HUS have become increasingly rare causes of CKD5 in children. CKD5 due to CM-HUS in the eculizumab era and due to STEC-HUS after improving supportive treatment is exceptional. Children on KRT due to STEC-HUS had a significantly better survival, shorter waiting time for kidney transplantation and better kidney graft survival compared to the CM-HUS group.
Collapse
Affiliation(s)
- Ilona Zagożdżon
- Department of Pediatrics, Nephrology and Hypertension, Medical University of Gdansk, Debinki 7, 80-211, Gdansk, Poland.
| | - Maria Szczepańska
- Department and Clinic of Pediatrics, Nephrology and Endocrinology, Medical University of Silesia, Zabrze, Poland
| | - Jacek Rubik
- Department of Nephrology and Kidney Transplantation, The Childrens Memorial Health Institute, Warsaw, Poland
| | - Katarzyna Zachwieja
- Department of Pediatric Nephrology and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| | - Anna Musielak
- Department of Pediatric Cardiology and Nephrology, Poznan University of Medical Sciences, Poznan, Poland
| | - Monika Bratkowska
- Department of Pediatrics, Immunology and Nephrology, Polish Mother's Memorial Hospital - Research Institute in Lodz, Lodz, Poland
| | - Irena Makulska
- Department of Pediatric Nephrology, Wroclaw Medical University, Wroclaw, Poland
| | - Katarzyna Niwińska
- Department of Pediatrics, Nephrology and Dialysis, Pomeranian Medical University, Szczecin, Poland
| | - Beata Leszczyńska
- Department of Pediatrics and Nephrology, Medical University of Warsaw, Warsaw, Poland
| | - Beata Bieniaś
- Department of Pediatric Nephrology, Medical University of Lublin, Lublin, Poland
| | | | | | - Aleksandra Żurowska
- Department of Pediatrics, Nephrology and Hypertension, Medical University of Gdansk, Debinki 7, 80-211, Gdansk, Poland
| |
Collapse
|
13
|
Sheth K, Lee C, Patel M, Mathew H, Saju A, Obligado S. Drug-Induced Thrombotic Microangiopathy Arising During the Treatment of Anal Carcinoma After the Use of Mitomycin C. Cureus 2025; 17:e81731. [PMID: 40330382 PMCID: PMC12050223 DOI: 10.7759/cureus.81731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2025] [Indexed: 05/08/2025] Open
Abstract
Anal cancer is a rare disease where malignant cells originate in the tissues of the anal canal. This form of cancer is classically treated with a combination of radiation therapy and a chemotherapy regimen that includes mitomycin C. This case illustrates an unusual presentation of thrombotic microangiopathy associated with mitomycin C. A 57-year-old woman with a history of anal carcinoma treated with capecitabine/mitomycin C and radiation was sent to the emergency department by her oncologist for an incidental finding of worsening kidney function noted on a complete metabolic panel done prior to getting radiographic imaging. The patient was admitted to the hospital for suspected acute kidney injury from suspected ureteral obstruction and stent occlusion; however, despite reversal of the stents, renal function did not improve. Renal biopsy confirmed thrombotic microangiopathy and diagnosis of drug-induced thrombotic microangiopathy. This case discusses a side effect of thrombotic microangiopathy from mitomycin C and successful treatment with eculizumab.
Collapse
Affiliation(s)
- Krishna Sheth
- Internal Medicine, Garnet Health Medical Center, Middletown, USA
| | - Cody Lee
- Internal Medicine, Touro College of Osteopathic Medicine, Middletown, USA
| | - Mihir Patel
- Emergency Medicine, University of Texas Southwestern Medical Center, Dallas, USA
| | - Hannah Mathew
- Internal Medicine, Garnet Health Medical Center, Middletown, USA
| | - Ajith Saju
- Internal Medicine, Garnet Health Medical Center, Middletown, USA
| | - Sergio Obligado
- Internal Medicine, Garnet Health Medical Center, Middletown, USA
| |
Collapse
|
14
|
Dreher L, Kuehl MB, Wenzel UO, Kylies D. Aortic aneurysm and dissection: complement and precision medicine in aortic disease. Am J Physiol Heart Circ Physiol 2025; 328:H814-H829. [PMID: 40019851 DOI: 10.1152/ajpheart.00853.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/08/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
Aortic disease encompasses life-threatening conditions such as aortic aneurysm and dissection, which are associated with high prevalence, morbidity, and mortality. The complement system, a key component of innate immunity, not only defends against pathogens but also maintains tissue homeostasis. Recent discoveries have expanded its role beyond immunity, linking complement dysregulation to numerous diseases and positioning it as a target for pharmacotherapy. Complement-based treatments for precision medicine are emerging, with several pharmaceuticals either already approved or under investigation. In aortic disease, complement activation and dysregulation have unveiled novel mechanisms and clinical implications. Human and experimental studies suggest that all three complement pathways contribute to disease pathophysiology. The complement system induces direct cellular damage via the membrane attack complex, as well as matrix-metalloproteinase (MMP)-associated tissue damage by promoting MMP-2 and MMP-9 expression. The anaphylatoxins C3a and C5a exacerbate disease by recruiting immune cells and triggering proinflammatory responses. Examples include neutrophil extracellular trap formation and cytokine release by polymorphonuclear neutrophils. These findings highlight the complement system as a promising novel diagnostic and therapeutic target in aortic disease with potential for individualized treatment. However, gaps remain, emphasizing the need for standardized multisite preclinical studies to improve reproducibility and translation. Biomarker studies must also be validated across diverse patient cohorts for clinical applicability. This review examines current knowledge regarding complement in aortic disease, aiming to evaluate its potential for innovative diagnostic and personalized treatment strategies.
Collapse
Affiliation(s)
- Leonie Dreher
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health, Hamburg, Germany
| | - Malte B Kuehl
- Department of Clinical Medicine - The Department of Pathology, Aarhus University, Aarhus, Denmark
| | - Ulrich O Wenzel
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health, Hamburg, Germany
| | - Dominik Kylies
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health, Hamburg, Germany
| |
Collapse
|
15
|
Musalem P. Ten tips for managing complement-mediated thrombotic microangiopathies (formerly atypical hemolytic uremic syndrome): narrative review. BMC Nephrol 2025; 26:158. [PMID: 40148764 PMCID: PMC11951749 DOI: 10.1186/s12882-025-04080-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Complement-mediated thrombotic microangiopathies (CM-TMA) are rare and life-threatening disorders characterized by microangiopathic hemolytic anemia, thrombocytopenia, and organ damage. These conditions result from dysregulation of the alternative complement pathway, often due to genetic variants or autoantibodies. The clinical spectrum is broad, comprising varied presentations and triggers, including infections, malignancies, and pregnancy-related complications. Advances in understanding the genetic and immunological basis of CM-TMA have improved diagnosis and treatment. Diagnosis requires exclusion of other thrombotic microangiopathies like thrombotic thrombocytopenic purpura and secondary causes, with genetic testing recommended to identify underlying susceptibilities. The introduction of C5 inhibitors has transformed the management of CM-TMA, significantly improving outcomes compared to the pre-2011 era when therapeutic plasma exchange was the primary therapy. Despite these advances, challenges remain in determining the optimal duration of therapy. Prophylactic measures against infections, particularly meningococcal disease, are mandatory for patients receiving C5 inhibitors. This article underscores the need for a personalized, multidisciplinary approach in the diagnosis and management of CM-TMA. Advances in genetics and complement biology have led to improved therapeutic strategies, however ongoing research is essential to address unanswered questions regarding relapse risk, treatment duration, and long-term outcomes.
Collapse
Affiliation(s)
- Pilar Musalem
- Nephrology, Dialysis and Transplantation Service, Hospital Las Higueras, Talcahuano, Concepción, Chile.
- Departamento de Medicina Interna, Facultad de Medicina, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
16
|
Kajana X, Caridi G, Bruschi M, Spinelli S, Lugani F, Ghiggeri GM, La Porta E, Mortari G, Verrina EE, Angeletti A, Bigatti C. The Crosstalk Between NETs and the Complement Cascade: An Overview in Nephrological Autoimmune Disease. Int J Mol Sci 2025; 26:2789. [PMID: 40141431 PMCID: PMC11943363 DOI: 10.3390/ijms26062789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
The complement cascade and Neutrophil Extracellular Traps (NETs) represent fundamental tools in protecting the host from foreign pathogens. Complement components and relative fragments, classically assigned to the innate immunity, represent a key link with the humoral immune response. NETs are a crucial component of the innate immune response, consisting of chromatin release from activated neutrophils. These web-like structures facilitate pathogen entrapment and elimination through proteolytic degradation and antimicrobial effectors. Previous findings suggested complement components and NETs have a significant role in the pathogenesis of several diseases characterized by inflammation, such as autoimmune and infectious diseases. However, the crosstalk between NETs and the complement cascade has only recently been investigated, and several aspects still need to be fully clarified. Recent evidence seems to suggest a bidirectional link between the complement cascade and NETosis. We here present the interaction between complement components and NETs in specific autoimmune diseases that mostly affect the kidney, such as systemic lupus erythematosus, Antineutrophilic Cytoplasmic Antibody (ANCA)-associated vasculitis and antiphospholipid syndrome. The mechanisms reported here may represent specific targets for the development of possible therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Andrea Angeletti
- Nephrology, Dialysis and Transplantation Unit, IRCCS Istituto Giannina Gaslini, 16145 Genoa, Italy; (X.K.); (G.C.); (M.B.); (S.S.); (F.L.); (G.M.G.); (E.L.P.); (G.M.); (E.E.V.); (C.B.)
| | | |
Collapse
|
17
|
Zanoni F, Obayemi JE, Gandla D, Castellano G, Keating BJ. Emerging role of genetics in kidney transplantation. Kidney Int 2025; 107:424-433. [PMID: 39710162 DOI: 10.1016/j.kint.2024.09.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 12/24/2024]
Abstract
The advent of more affordable genomic analytical pipelines has facilitated the expansion of genetic studies in kidney transplantation. Advances in genetic sequencing have allowed for a greater understanding of the genetic basis of chronic kidney disease, which has helped to guide transplant management and address issues related to living donation in specific disease settings. Recent efforts have shown significant effects of genetic ancestry and donor APOL1 risk genotypes in determining worse allograft outcomes and increased donation risks. Genetic studies in kidney transplantation outcomes have started to assess the effects of donor and recipient genetics in primary disease recurrence and transplant-related comorbidities, while genome-wide donor-recipient genetic incompatibilities have been shown to represent an important determinant of alloimmunity. Future large-scale comprehensive studies will shed light on the clinical utility of integrative genomics in the kidney transplantation setting.
Collapse
Affiliation(s)
- Francesca Zanoni
- Department of Nephrology, Dialysis and Kidney Transplantation, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Division of Transplantation, Department of Surgery, New York University Langone Health, Grossman School of Medicine, New York, New York, USA
| | - Joy E Obayemi
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA; Comprehensive Transplant Center, Department of Surgery, Northwestern University, Chicago Illinois, USA
| | - Divya Gandla
- Division of Transplantation, Department of Surgery, New York University Langone Health, Grossman School of Medicine, New York, New York, USA
| | - Giuseppe Castellano
- Department of Nephrology, Dialysis and Kidney Transplantation, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Brendan J Keating
- Institute of Systems Genetics, New York University Langone Health, Grossman School of Medicine, New York, New York, USA.
| |
Collapse
|
18
|
Kano T, Io H, Sasaki Y, Muto M, Muto S, Ogiwara K, Ikeda A, Iwasaki H, Suzuki Y. A Case of Atypical Hemolytic Uremic Syndrome With a Complement Factor I Mutation Triggered by a Femoral Neck Fracture. Nephrology (Carlton) 2025; 30:e70010. [PMID: 40001340 PMCID: PMC11861886 DOI: 10.1111/nep.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
Atypical hemolytic uremic syndrome is a thrombotic microangiopathy caused by the abnormal activation of the alternative complement pathway. Mutations in complement-related genes and autoantibodies against complement regulators are involved in the pathogenesis of this condition; the frequency of, and prognosis of patients harbouring, each genetic mutation varies based on the region and race. Complement factor I (CFI) mutations have been observed in 4%-8% of cases in Europe; however, they have not yet been reported in Japan. We present the first Japanese case of atypical hemolytic uremic syndrome in a patient harbouring a CFI mutation. An 83-year-old female patient presented with severe acute kidney injury, thrombocytopenia, and hemolytic anaemia following a femoral neck fracture. Plasma exchange and haemodialysis were initiated, resulting in improved kidney function and platelet count. However, the platelet count decreased when plasma exchange was discontinued. Therefore, we administered ravulizumab, an anti-complement 5 monoclonal antibody, which led to the maintenance of stable kidney function and platelet count. Genetic analysis revealed a CFI mutation, and the patient was treated with ravulizumab for 2 years without relapse. Individuals diagnosed with atypical hemolytic uremic syndrome harbouring CFI mutations experience poor outcomes, including low rates of remission, high rates of mortality, and progression to end-stage kidney disease. Our case serves as a crucial example demonstrating how prompt identification and appropriate management can lead to better patient outcomes.
Collapse
Affiliation(s)
- Toshiki Kano
- Department of NephrologyJuntendo University Nerima HospitalTokyoJapan
- Department of NephrologyJuntendo University Faculty of MedicineTokyoJapan
| | - Hiroaki Io
- Department of NephrologyJuntendo University Nerima HospitalTokyoJapan
| | - Yu Sasaki
- Department of NephrologyJuntendo University Nerima HospitalTokyoJapan
| | - Masahiro Muto
- Department of NephrologyJuntendo University Nerima HospitalTokyoJapan
- Department of NephrologyJuntendo University Faculty of MedicineTokyoJapan
| | - Sayaka Muto
- Department of NephrologyJuntendo University Nerima HospitalTokyoJapan
- Department of NephrologyJuntendo University Faculty of MedicineTokyoJapan
| | - Kei Ogiwara
- Department of NephrologyJuntendo University Nerima HospitalTokyoJapan
- Department of NephrologyJuntendo University Faculty of MedicineTokyoJapan
| | - Arisa Ikeda
- Department of NephrologyJuntendo University Nerima HospitalTokyoJapan
- Department of NephrologyJuntendo University Faculty of MedicineTokyoJapan
| | - Hiroyuki Iwasaki
- Department of NephrologyJuntendo University Nerima HospitalTokyoJapan
- Department of NephrologyJuntendo University Faculty of MedicineTokyoJapan
| | - Yusuke Suzuki
- Department of NephrologyJuntendo University Faculty of MedicineTokyoJapan
| |
Collapse
|
19
|
Jang JH, Han B, Jung J, Russo P, Kulasekararaj AG. The Path to Accessible Care: Development and Impact of Eculizumab Biosimilars for Paroxysmal Nocturnal Hemoglobinuria and Atypical Hemolytic Uremic Syndrome. BioDrugs 2025; 39:281-295. [PMID: 39982653 PMCID: PMC11906501 DOI: 10.1007/s40259-025-00707-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2025] [Indexed: 02/22/2025]
Abstract
Eculizumab, a humanized monoclonal antibody targeting complement C5, is the first approved drug for complement-mediated diseases and indicated to treat paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, myasthenia gravis, and neuromyelitis optica spectrum disorder. The introduction of eculizumab has improved the prognosis of paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome to near-normal life expectancy and quality of life. Administration of eculizumab resulted in a rapid and sustained reduction in hemolytic activity and a consequent risk of thrombosis in paroxysmal nocturnal hemoglobinuria, and thrombotic microangiopathy in atypical hemolytic uremic syndrome, respectively. Nevertheless, many patients still have difficulty accessing eculizumab treatment because of its high costs. Biosimilars to reference eculizumab may increase patient access to treatment by creating market competition and eventually decreasing treatment costs. Clinical use of biosimilars in Europe in the last 15 years has demonstrated that they are as safe and effective as their reference products, and can also drive cost reductions and increase patients' access to treatment. This review aims to increase awareness about the importance of biosimilars of reference eculizumab and their entry for use in patients with paroxysmal nocturnal hemoglobinuria or atypical hemolytic uremic syndrome based on the accumulated experience of other previously approved biosimilars, and to provide an overview of the stringent biosimilar development pathway in general and the concept of extrapolation in particular.
Collapse
Affiliation(s)
- Jun Ho Jang
- Department of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
| | - Bing Han
- Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Jinah Jung
- Samsung Bioepis Co., Ltd., Incheon, Republic of Korea
| | - Paola Russo
- Samsung Bioepis Co., Ltd., Incheon, Republic of Korea
| | - Austin G Kulasekararaj
- King's College Hospital-NHS Foundation Trust, NIHR/Wellcome King's Clinical Research Facility, and King's College London, London, UK
| |
Collapse
|
20
|
Ter Avest M, Duineveld C, Bouwmeester RN, Baas LM, van den Heuvel LPWJ, Langemeijer SMC, Wetzels JFM, van de Kar NCAJ, Ter Heine R. Prospective validation of initial eculizumab dosing in adults with atypical hemolytic uremic syndrome. Nephrol Dial Transplant 2025; 40:598-601. [PMID: 39533120 DOI: 10.1093/ndt/gfae244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Indexed: 11/16/2024] Open
Affiliation(s)
- Mendy Ter Avest
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
| | - Caroline Duineveld
- Department of Nephrology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Romy N Bouwmeester
- Department of Pediatric Nephrology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, Netherlands
| | - Laura M Baas
- Department of Pediatric Nephrology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, Netherlands
| | - Lambertus P W J van den Heuvel
- Department of Pediatric Nephrology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Pediatrics/Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, University Hospitals Leuven, Leuven, Belgium
| | | | - Jack F M Wetzels
- Department of Nephrology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Nicole C A J van de Kar
- Department of Pediatric Nephrology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rob Ter Heine
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
21
|
Mohamud Y, Bahreyni A, Hwang SW, Lin JC, Wang ZC, Zhang J, Luo H. Mitochondrial injury and complement dysregulation are drivers of pathological inflammation in viral myocarditis. J Virol 2025; 99:e0180424. [PMID: 39846741 PMCID: PMC11852726 DOI: 10.1128/jvi.01804-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/11/2024] [Indexed: 01/24/2025] Open
Abstract
Enteroviruses cause nearly 1 billion global infections annually and are associated with a diverse array of human illnesses. Among these, myocarditis and the resulting chronic inflammation have been recognized as major contributing factors to virus-induced heart failure. Despite our growing understanding, very limited therapeutic strategies have been developed to address the pathological consequences of virus-induced chronic innate immune activation. Coxsackievirus B3 (CVB3) was used as a model cardiotropic enterovirus. We leveraged in vitro cell-based studies to investigate cardiomyocyte and macrophage interaction during CVB3 infection, as well as animal studies and unique human cardio specimens to evaluate mechanisms of viral heart injury. We present evidence that viral myocarditis is in part exacerbated by pathological activation of the complement pathway in cells, mice, and human cardiac tissues. We demonstrate unique cell type-specific responses to viral infection that are exacerbated by mitochondrial injury in cardiomyocytes and NFκB-dependent pro-inflammatory response in macrophages. Macrophages are robustly activated by damage-associated mitochondrial components, including mitochondrial proteins and lipid extracts. Mechanistically, we identify complement protective factors CD59/protectin and CD55/DAF as novel targets of viral proteinase that acts to release the brakes on complement-mediated autoinjury. Collectively, our study highlights a novel mechanism that can act as a potential contributor to CVB3 pathogenesis through mitochondrial injury-mediated autoimmunity. IMPORTANCE This study sheds light on how enteroviruses, specifically coxsackievirus B3, may contribute to heart failure by triggering harmful immune responses in the heart. We discovered that viral infections in heart cells cause mitochondrial damage, which in turn activates a destructive immune response involving the complement system. This immune activation is one of the significant contributors that lead to further injury of heart tissues, worsening the damage caused by the virus. Additionally, we identified key protective molecules that are targeted and disrupted by the virus, allowing the immune system to attack the heart even more aggressively. Understanding these mechanisms may provide additional insights into how viral infections can lead to chronic heart conditions and suggests potential therapeutic targets to prevent or reduce heart damage in patients affected by viral myocarditis.
Collapse
Affiliation(s)
- Yasir Mohamud
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amirhossein Bahreyni
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sinwoo Wendy Hwang
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jingfei Carly Lin
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zhihan Claire Wang
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jingchun Zhang
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
22
|
Duneton C, Kwon T, Dossier C, Baudouin V, Fila M, Mariani-Kurkdijan P, Nel I, Boyer O, Hogan J. IgG-immunoadsorptions and eculizumab combination in STEC-hemolytic and uremic syndrome pediatric patients with neurological involvement. Pediatr Nephrol 2025; 40:431-440. [PMID: 39297957 DOI: 10.1007/s00467-024-06418-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/08/2024] [Accepted: 05/06/2024] [Indexed: 09/21/2024]
Abstract
BACKGROUND Neurological complications pose a significant threat in pediatric hemolytic and uremic syndrome (HUS) resulting from infections with Shiga toxin-producing Escherichia coli (STEC), with no established treatment. The involvement of complement activation in the pathogenesis of STEC-HUS is acknowledged, and eculizumab (ECZ), a terminal complement blocker, has been documented in several pediatric series with inconsistent results. Antibody-mediated mechanisms have also been suggested, with IgG-immunoadsorption (IgIA) showing promise in adults with neurological complications. We aimed to assess the benefit of combining IgIA with ECZ in pediatric patients with neurological STEC-HUS compared to patients treated with ECZ alone or supportive care. METHODS Multicenter retrospective study conducted on pediatric patients (< 18 years) with neurological STEC-HUS treated with IgIA + ECZ or ECZ alone from 2010 to 2020 in France. A historical cohort treated with supportive care served as controls. Primary outcome included survival and neurological evaluation at 1-year follow-up (dichotomized as normal vs. abnormal). RESULTS A total of 42 children were included: 18 treated with IgIA + ECZ, 24 with ECZ alone, and 27 with supportive care. Although there was no significant difference in survival between groups, three deaths occurred in the control group in the acute phase, while none was reported in both the IgIA + ECZ and ECZ alone groups, despite presenting with more severe neurological symptoms for IgIA + ECZ patients. No significant association was found between treatment group and 1-year neurological evaluation after adjustment for age, sex, and initial neurological presentation. CONCLUSIONS Systematic association of IgIA + ECZ is not supported for all neurological STEC-HUS pediatric patients; potential rescue therapy for severe cases warrants consideration.
Collapse
Affiliation(s)
- Charlotte Duneton
- Pediatric Nephrology, Dialysis and Transplantation Department, Robert Debré University Hospital, APHP, Paris Cité University, Paris, France.
- Université Paris Cité, INSERM U976, HIPI Unit: Human Immunology, Pathology, Immunotherapy, Paris, France.
| | - Theresa Kwon
- Pediatric Nephrology, Dialysis and Transplantation Department, Robert Debré University Hospital, APHP, Paris Cité University, Paris, France
| | - Claire Dossier
- Pediatric Nephrology, Dialysis and Transplantation Department, Robert Debré University Hospital, APHP, Paris Cité University, Paris, France
| | - Veronique Baudouin
- Pediatric Nephrology, Dialysis and Transplantation Department, Robert Debré University Hospital, APHP, Paris Cité University, Paris, France
| | - Marc Fila
- Pediatric Nephrology Department, Montpellier University Hospital, Montpellier, France
| | - Patricia Mariani-Kurkdijan
- Department of Microbiology, National Reference Center for Escherichia Coli, Robert Debré University Hospital, APHP, Paris Cité University, Paris, France
| | - Isabelle Nel
- Immunology Laboratory, Robert Debré University Hospital, APHP, Paris Cité University, Paris, France
- Université Paris Cité, INSERM U976, HIPI Unit: Human Immunology, Pathology, Immunotherapy, Paris, France
| | - Olivia Boyer
- Pediatric Nephrology Department, CNR-MAT, Imagine Institute, Necker University Hospital, APHP, Paris Cité University, Paris, France
| | - Julien Hogan
- Pediatric Nephrology, Dialysis and Transplantation Department, Robert Debré University Hospital, APHP, Paris Cité University, Paris, France
- Université Paris Cité, INSERM, UMR-S970, PARCC, Paris Translational Research Center for Organ Transplantation, Paris, France
| |
Collapse
|
23
|
Gäckler A, Brinkkötter PT. [Atypical hemolytic uremic syndrome: differential diagnosis and therapy - A clinical practice guideline for diagnosis and therapy]. Dtsch Med Wochenschr 2025; 150:167-172. [PMID: 39879972 DOI: 10.1055/a-2382-6433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Atypical hemolytic uremic syndrome (aHUS) is an important differential diagnosis in thrombotic microangiopathy (TMA). The absence of definitive biomarkers usually allows for aHUS to be diagnosed only through a process of exclusion. Due to the unfavorable prognosis if adequate therapy is delayed or not provided, differential diagnostic considerations and initiation of treatment must occur promptly. The presented guideline is intended to serve as an aid in this process.
Collapse
|
24
|
Meena P, Gala R, Das RR, Bhargava V, Saivani Y, Panda S, Mantri A, Agrawaal KK. Kidney and pregnancy outcomes in pregnancy-associated atypical hemolytic uremic syndrome: A systematic review and meta-analysis. Medicine (Baltimore) 2025; 104:e41403. [PMID: 39889176 PMCID: PMC11789862 DOI: 10.1097/md.0000000000041403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 02/02/2025] Open
Abstract
BACKGROUND Pregnancy-associated atypical hemolytic uremic syndrome (p-aHUS) is a rare, life-threatening condition characterized by microangiopathic hemolytic anemia, thrombocytopenia, elevated liver enzymes, and acute kidney injury. Prompt diagnosis and therapy are crucial due to the high risk of progression to chronic kidney disease (CKD), end-stage kidney disease (ESKD), and dialysis dependency, as well as significant maternal and fetal morbidity and mortality. METHODS A comprehensive literature search was conducted across EMBASE, MEDLINE, and the Cochrane CENTRAL from January 2000 to March 2024. Studies reporting on pregnancy and kidney outcomes in women diagnosed with p-aHUS were included. RESULTS Ten studies involving 386 pregnancies in 380 patients met the inclusion criteria for the final analysis. Renal outcomes varied, with mean creatinine levels ranging from 0.72 to 8.734 mg/dL. Dialysis was required in 66.6% of patients, and 25% developed ESKD. Maternal complications included preeclampsia (36.4%) and hemolysis, elevated liver enzymes, and low platelets syndrome (29.7%), with a 5% maternal mortality rate. Fetal complications included intrauterine fetal demise (n = 25), intrauterine growth restriction, low birth weight, and prematurity. Treatment with eculizumab significantly reduced the risk of CKD and ESKD, with a pooled risk ratio of 0.20 (95% confidence interval: 0.09-0.44) and low heterogeneity (I² = 0%, P = .43). CONCLUSION This analysis highlights the severe kidney and pregnancy outcomes associated with p-aHUS. Eculizumab treatment is significantly beneficial in reducing the risk of CKD and ESKD.
Collapse
Affiliation(s)
- Priti Meena
- Department of Nephrology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Ruju Gala
- Department of Nephrology, Zynova Shalby Hospital, Mumbai, Maharashtra, India
| | - Rashmi Ranjan Das
- Department of Pediatrics, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Vinant Bhargava
- Department of Nephrology, Sir Ganga Ram Hospital, New Delhi, India
| | - Yellampalli Saivani
- Department of Nephrology, Santhiram Medical College, Nadyal, Andhra Pradesh, India
| | - Sandip Panda
- Department of Nephrology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Alok Mantri
- Department of Gastroenterology, Kalinga Hospital, Bhubaneswar, Odisha, India
| | - Krishna Kumar Agrawaal
- Department of Nephrology, Universal College of Medical Sciences, Bhairahawa, Siddhartha Nagar, Nepal
| |
Collapse
|
25
|
Kurts C, von Vietinghoff S, Krebs CF, Panzer U. Kidney immunology from pathophysiology to clinical translation. Nat Rev Immunol 2025:10.1038/s41577-025-01131-y. [PMID: 39885266 DOI: 10.1038/s41577-025-01131-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2025] [Indexed: 02/01/2025]
Abstract
Kidney diseases are widespread and represent a considerable medical, social and economic burden. However, there has been marked progress in understanding the immunological aspects of kidney disease. This includes the identification of distinct intrarenal immunological niches and characterization of kidney disease endotypes according to the underlying molecular immunopathology, as well as a better understanding of the pathological roles for T cells, mononuclear phagocytes and B cells and the renal elements they target. These insights have improved the diagnosis of kidney disease. Here, we discuss new developments in our understanding of kidney immunology, focusing on immune mechanisms of disease and their translational implications for the diagnosis and treatment of kidney disease. We also describe the immune-mediated crosstalk between the kidney and other organs that influences kidney disease and extrarenal inflammation.
Collapse
Affiliation(s)
- Christian Kurts
- Institute of Molecular Medicine and Experimental Immunology, University Hospital, Bonn, Germany.
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia.
| | - Sibylle von Vietinghoff
- Nephrology Section, University Hospital Bonn, Medical Clinic and Polyclinic I, Bonn, Germany
| | - Christian F Krebs
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulf Panzer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
26
|
Gentile M, Manenti L. Targeted Complement Treatments in Glomerulopathies: A Comprehensive Review. J Clin Med 2025; 14:702. [PMID: 39941374 PMCID: PMC11818541 DOI: 10.3390/jcm14030702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
The complement system includes soluble and cell surface proteins and is an important arm of the innate immune system. Once activated, the complement system rapidly generates proteins with inflammatory and vasoactive activities. Although complement is crucial to host defense and homeostasis, its inappropriate or uncontrolled activation can also drive tissue injury. Glomerulopathy encompasses a spectrum of diseases with diverse etiologies, clinical presentations, and outcomes. Among the intricate web of factors contributing to glomerulopathies pathogenesis, the role of complement activation has emerged as a focal point of research interest and therapeutic intervention. The pioneer drug was eculizumab, which made it possible to drastically change the prognosis of atypical hemolytic uremic syndrome, an otherwise fatal disease. This comprehensive review aims to elucidate the multifaceted interplay between complement pathways and glomerulopathies, shedding light on potential pathways for targeted therapies and improved patient care.
Collapse
Affiliation(s)
- Micaela Gentile
- UO Nefrologia, Dipartimento di Medicina e Chirurgia, Università di Parma, 43126 Parma, Italy;
| | - Lucio Manenti
- Nephrology Unit, Azienda Sociosanitaria Liguria 5, 19121 La Spezia, Italy
| |
Collapse
|
27
|
Kopland M, Ottersen VS, Dahle DO, Gedde-Dahl T, Dalgaard J. En mann i 50-årene med akutt nyresvikt, anemi og trombocytopeni. TIDSSKRIFT FOR DEN NORSKE LEGEFORENING 2025; 145:24-0231. [PMID: 39835860 DOI: 10.4045/tidsskr.24.0231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Abstract
Background When haemolytic anaemia, thrombocytopenia and renal failure are present, a thrombotic microangiopathic (TMA) condition should be suspected. We describe the various differential diagnoses of primary TMA syndromes, their clinical findings, clinical workup and treatment. Case presentation A previously healthy man in his fifties was hospitalised with anaemia, thrombocytopenia, bilirubinaemia and acute renal failure. He was in good general condition without neurological symptoms. Due to suspicion of thrombotic microangiopathy (mainly thrombotic thrombocytopenic purpura (TTP)), he was referred to a university clinic for immediate plasma exchange. His renal failure deteriorated and haemodialysis was initiated. However, ADAMTS13 level was normal. Thus, complement-mediated thrombotic microangiopathy was suspected and eculizumab was administered. Due to previous abdominal discomfort a faecal sampling was performed, detecting enterohaemorrhagic E. coli (EHEC) DNA (Shigatoxin 2f). Hence, the diagnosis was Shigatoxin-induced haemolytic uraemic syndrome (ST-HUS). Supportive care was continued and renal function normalised. Interpretation When thrombotic microangiopathy is suspected, an assessment is made whether to initiate plasmapheresis and immunomodulatory treatment. A thorough workup is needed to differentiate between the various primary TMA syndromes. Thus, treatment with expensive and possibly harmful drugs can be avoided. The various primary TMA syndromes have overlapping clinical features, and the diagnosis can be difficult.
Collapse
Affiliation(s)
| | | | - Dag Olav Dahle
- Seksjon for nyremedisin, Avdeling for transplantasjonsmedisin, Oslo universitetssykehus
| | - Tobias Gedde-Dahl
- Avdeling for blodsykdommer, Oslo universitetssykehus, og, Institutt for klinisk medisin, Universitetet i Oslo
| | | |
Collapse
|
28
|
Lokkur P, Bansal SB. Complement in Kidney Transplantation. Transplant Rev (Orlando) 2025; 39:100897. [PMID: 39615219 DOI: 10.1016/j.trre.2024.100897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/13/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025]
Abstract
Transplantation is the treatment of choice in most patients with kidney failure. The complement system plays a vital role in transplantation. The complement system forms a major part of innate immunity and acts as a bridge between innate and acquired immunity. Many diseases, particularly concerning the kidneys, result from complement system dysregulation, like atypical hemolytic uremic syndrome (aHUS), C3 glomerulopathy (C3GN), systemic lupus erythematosus (SLE and some other immune complex diseases. The complement system activation is a very important part of post-transplant events like ischemia-reperfusion injury (IRI), delayed graft function (DGF), antibody-mediated rejection (ABMR) and thrombotic microangiopathy (TMA). A better understanding of the complement cascade can help to plan strategies to prevent and manage complement-related problems before and after kidney transplantation. Many newer molecules are either being developed or in the pipeline, which target the complement system at various stages. These novel therapeutics are now considered additional measures to improve graft survival. This review summarises the complement cascade, its role in kidney diseases and kidney transplantation, and possible areas of target and novel therapeutics.
Collapse
Affiliation(s)
- Pooja Lokkur
- Department of Nephrology and Kidney Transplantation, Medanta Medicity, Sector 38, Gurgaon 122001, India
| | - Shyam Bihari Bansal
- Department of Nephrology and Kidney Transplantation, Medanta Medicity, Sector 38, Gurgaon 122001, India.
| |
Collapse
|
29
|
Cole MA, Ranjan N, Gerber GF, Pan XZ, Flores-Guerrero D, McNamara G, Chaturvedi S, Sperati CJ, McCrae KR, Brodsky RA. Complement biosensors identify a classical pathway stimulus in complement-mediated thrombotic microangiopathy. Blood 2024; 144:2528-2545. [PMID: 39357054 PMCID: PMC11862816 DOI: 10.1182/blood.2024025850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/23/2024] [Accepted: 09/13/2024] [Indexed: 10/04/2024] Open
Abstract
ABSTRACT Complement-mediated thrombotic microangiopathy (CM-TMA) or hemolytic uremic syndrome, previously identified as atypical hemolytic uremic syndrome, is a TMA characterized by germ line variants or acquired antibodies to complement proteins and regulators. Building upon our prior experience with the modified Ham (mHam) assay for ex vivo diagnosis of complementopathies, we have developed an array of cell-based complement "biosensors" by selective removal of complement regulatory proteins (CD55 and CD59, CD46, or a combination thereof) in an autonomously bioluminescent HEK293 cell line. These biosensors can be used as a sensitive method for diagnosing CM-TMA and monitoring therapeutic complement blockade. Using specific complement pathway inhibitors, this model identifies immunoglobulin M (IgM)-driven classical pathway stimulus during both acute disease and in many patients during clinical remission. This provides a potential explanation for ∼50% of patients with CM-TMA who lack an alternative pathway "driving" variant and suggests at least a subset of CM-TMA is characterized by a breakdown of IgM immunologic tolerance.
Collapse
Affiliation(s)
- Michael A. Cole
- Division of Hematology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Nikhil Ranjan
- Division of Hematology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Gloria F. Gerber
- Division of Hematology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Xiang-Zuo Pan
- Division of Hematology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | | | - George McNamara
- Ross Fluorescence Imaging Center, Division of Gastroenterology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Shruti Chaturvedi
- Division of Hematology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - C. John Sperati
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Keith R. McCrae
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Robert A. Brodsky
- Division of Hematology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
30
|
Java A. Atypical hemolytic uremic syndrome: diagnosis, management, and discontinuation of therapy. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2024; 2024:200-205. [PMID: 39644051 DOI: 10.1182/hematology.2024000543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
Atypical hemolytic uremic syndrome (aHUS) is a thrombotic microangiopathy typically characterized by anemia, thrombocytopenia, and end-organ injury. aHUS occurs due to endothelial injury resulting from overactivation of the alternative pathway of the complement system. The etiology of the dysregulated complement system is either a genetic mutation in 1 or more complement proteins or an acquired deficiency due to autoantibodies. Over the past decade, advancements in our understanding of the role of complement in the pathophysiology of aHUS as well as the availability of anticomplement drugs has been a game-changer for our patients. These drugs have revolutionized the clinical course, outcome, and prognosis of this disease. Therefore, all patients in whom aHUS is suspected should undergo testing for complement genetic variants and autoantibodies. In approximately 30% to 40% of patients, a genetic variant of uncertain significance (VUS) may be identified. Such patients should undergo further testing to define the significance of the VUS. A combination of antigenic, functional, and biomarker analyses can assist in establishing the significance of the variants and thereby define the etiology in most patients. These analyses will also help to determine the duration of treatment based on the individual's genetic alteration. This review aims to shed light on the diagnosis and management of aHUS and discusses how to stratify patients to determine who can safely discontinue anticomplement therapy.
Collapse
Affiliation(s)
- Anuja Java
- Department of Medicine, Division of Nephrology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
31
|
Kavanagh D, Ardissino G, Brocklebank V, Bouwmeester RN, Bagga A, Ter Heine R, Johnson S, Licht C, Ma ALT, Noris M, Praga M, Rondeau E, Sinha A, Smith RJH, Sheerin NS, Trimarchi H, Wetzels JFM, Vivarelli M, Van de Kar NCAJ, Greenbaum LA. Outcomes from the International Society of Nephrology Hemolytic Uremic Syndromes International Forum. Kidney Int 2024; 106:1038-1050. [PMID: 39395628 DOI: 10.1016/j.kint.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/07/2024] [Accepted: 09/04/2024] [Indexed: 10/14/2024]
Abstract
Hemolytic uremic syndromes (HUSs) are a heterogeneous group of conditions, only some of which are mediated by complement (complement-mediated HUS). We report the outcome of the 2023 International Society of Nephrology HUS International Forum where a global panel of experts considered the current state of the art, identified areas of uncertainty, and proposed optimal solutions. Areas of uncertainty and areas for future research included the nomenclature of HUS, novel complement testing strategies, identification of biomarkers, genetic predisposition to atypical HUS, optimal dosing and withdrawal strategies for C5 inhibitors, treatment of kidney transplant recipients, disparity of access to treatment, and the next generation of complement inhibitors in complement-mediated HUS. The current rationale for optimal patient management is described.
Collapse
Affiliation(s)
- David Kavanagh
- National Renal Complement Therapeutics Centre, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, Newcastle upon Tyne, United Kingdom; Complement Therapeutics Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | - Gianluigi Ardissino
- Center for Hemolytic Uremic Syndrome (HUS) Prevention, Pediatric Nephrology, Dialysis and Transplantation Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Vicky Brocklebank
- National Renal Complement Therapeutics Centre, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, Newcastle upon Tyne, United Kingdom; Complement Therapeutics Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Romy N Bouwmeester
- Radboud University Medical Center, Amalia Children's Hospital, Radboud Institute for Molecular Life Sciences, Department of Pediatric Nephrology, Nijmegen, The Netherlands
| | - Arvind Bagga
- Division of Nephrology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Rob Ter Heine
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sally Johnson
- National Renal Complement Therapeutics Centre, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, Newcastle upon Tyne, United Kingdom; Great North Children's Hospital, Sir James Spence Institute, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Christoph Licht
- Division of Nephrology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Alison L T Ma
- Paediatric Nephrology Centre, Hong Kong Children's Hospital, Hong Kong, China; Department of Pediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Marina Noris
- Istituto di Ricerche Farmacologiche Mario Negri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Clinical Research Center for Rare Diseases Aldo e Cele Daccò, Ranica, Italy
| | - Manuel Praga
- Department of Nephrology, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain; Medicine Department, Universidad Complutense de Madrid, Madrid, Spain
| | - Eric Rondeau
- Intensive Care Nephrology and Transplantation Department, Assistance Publique des Hôpitaux de Paris (AP-HP) - Sorbonne Université, Paris, France
| | - Aditi Sinha
- Division of Nephrology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Richard J H Smith
- Molecular Otolaryngology and Renal Research Laboratories, The University of Iowa, Iowa City, Iowa, USA
| | - Neil S Sheerin
- National Renal Complement Therapeutics Centre, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, Newcastle upon Tyne, United Kingdom; Complement Therapeutics Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - H Trimarchi
- Nephrology Service, Hospital Britanico de Buenos Aires, Buenos Aires, Argentina
| | - Jack F M Wetzels
- Department of Nephrology, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Marina Vivarelli
- Division of Nephrology, Laboratory of Nephrology, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Nicole C A J Van de Kar
- Radboud University Medical Center, Amalia Children's Hospital, Radboud Institute for Molecular Life Sciences, Department of Pediatric Nephrology, Nijmegen, The Netherlands
| | - Larry A Greenbaum
- Division of Pediatric Nephrology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA.
| |
Collapse
|
32
|
Duan H, Zhang Y, Otis MR, Drolet DW, Geisbrecht BV. The Inhibitory Effects of a Factor B-Binding DNA Aptamer Family Supersede the Gain of Function of Factor B Variants Associated with Atypical Hemolytic Uremic Syndrome. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1691-1702. [PMID: 39431879 PMCID: PMC11573645 DOI: 10.4049/jimmunol.2400420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/30/2024] [Indexed: 10/22/2024]
Abstract
Aptamers are short, single-stranded oligonucleotides that selectively bind to target biomolecules. Although they generally exhibit good binding specificity, their affinities are often limited because of the relative lack of hydrophobic groups in nucleic acids. Chemically modified nucleotides incorporating hydrophobic structures into uracil have been synthesized to address this obstacle. Modified DNA aptamers containing such nonstandard nucleotides have been developed for >20 different complement proteins. These modified aptamers show increased affinity and enhanced serum stability and have potential value as therapeutic agents. We recently conducted a structure/function study on a family of modified DNA aptamers that bind specifically to complement Factor B (FB). This work revealed that these aptamers selectively inhibit the complement alternative pathway (AP) by preventing the formation of the AP complement component C3 (C3) proconvertase complex, C3bB. Certain patients with atypical hemolytic uremic syndrome express gain-of-function variants of FB that enhance the formation of the proconvertase complex and/or decrease the efficacy of endogenous regulators against the C3 convertases they form. To investigate whether these FB-binding aptamers could override the effects of disease-causing mutations in FB, we examined how they interacted with several FB variants, including D279G, F286L, K323E, and K350N, in various assays of complement function. We found that the inhibitory effect of the FB-binding aptamers superseded the gain-of-function mutations in FB, although the aptamers could not dissociate preformed C3 convertases. These findings suggest that FB-binding aptamers could be further developed as a potential treatment for certain atypical hemolytic uremic syndrome patients or those with other diseases characterized by excessive complement activity.
Collapse
Affiliation(s)
- Huiquan Duan
- Department of Biochemistry & Molecular Biophysics, Kansas State University; Manhattan, KS 66506 U.S.A
| | - Ying Zhang
- Department of Biochemistry & Molecular Biophysics, Kansas State University; Manhattan, KS 66506 U.S.A
| | | | | | - Brian V. Geisbrecht
- Department of Biochemistry & Molecular Biophysics, Kansas State University; Manhattan, KS 66506 U.S.A
| |
Collapse
|
33
|
Vellanki S, Trikannad Ashwini Kumar AK, Thalambedu N, Gentille C, Varma A. Eculizumab for Hyperhemolysis in Sickle Cell Disease: Key Insights From a Challenging Case. ANNALS OF INTERNAL MEDICINE: CLINICAL CASES 2024; 3. [DOI: 10.7326/aimcc.2024.0424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Affiliation(s)
- Sruthi Vellanki
- Department of Hematology-Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | - Nishanth Thalambedu
- Department of Hematology-Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Cesar Gentille
- Department of Hematology-Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Ankur Varma
- Department of Hematology-Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
34
|
Ryder S. Integrated Applied Clinical Pharmacology in the Advancement of Rare and Ultra-Rare Disease Therapeutics. Clin Pharmacol Ther 2024; 116:1485-1495. [PMID: 39034754 DOI: 10.1002/cpt.3382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/29/2024] [Indexed: 07/23/2024]
Abstract
The introduction of safe and effective rare/ultra-rare disease treatments is a focus of many biotherapeutic enterprises. Despite this increased activity, a significant unmet need remains, and the responsibility to meet this need is augmented by enhanced genomic, biologic, medical, analytical, and informatic tools. It is recognized that the development of an effective and safe rare/ultra-rare disease therapeutic faces a number of challenges with an important role noted for clinical pharmacology. Clinical pharmacology is foundationally an integrative discipline which must be embedded in and is interdependent upon understanding the pathogenic biology, clinical presentation, disease progression, and end-point assessment of the disease under study. This manuscript presents an overview and two case examples of this integrative approach, the development of C5-targeted therapeutics for the treatment of generalized myasthenia gravis and asfotase alpha for the treatment of hypophosphatasia. The two presented case examples show the usefulness of understanding the biological drivers and clinical course of a rare disease, having relevant animal models, procuring informative natural history data, importing assessment tools from relevant alternative areas, and using integrated applied clinical pharmacology to inform target engagement, dose, and the cascade of pharmacodynamic and clinical effects that follow. Learnings from these programs include the importance of assuring cross-validation of assays throughout a development program and continued commitment to understanding the relationship among the array of Pd end points and clinical outcomes. Using an integrative approach, substantive work remains to be done to meet the unmet needs of patients with rare/ultra-rare disease.
Collapse
|
35
|
Joseph A, Harel S, Mesnard L, Rafat C, Knapp S, Rumpler A, Philipponnet C, Barba C, Rebibou JM, Buob D, Hertig A, Vargaftig J, Halimi JM, Arnulf B, Bretaud AS, Joly B, Grangé S, Coppo P. Carfilzomib-associated thrombotic microangiopathy: clinical features and outcomes. Nephrol Dial Transplant 2024; 39:2067-2078. [PMID: 38658194 DOI: 10.1093/ndt/gfae096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Carfilzomib, a new proteasome inhibitor indicated for patients with relapsed/refractory myeloma, has been associated with cases of thrombotic microangiopathy (CFZ-TMA). The role of variants in the complement alternative pathway and therapeutic potential of complement blockade with eculizumab remain to be determined. METHODS We report 37 cases of CFZ-TMA recorded in the French reference center for TMA with their clinical characteristics, genetic analysis and outcome according to treatments. RESULTS A trigger was identified in more than half of cases, including eight influenza and five severe acute respiratory syndrome coronavirus-2 cases. All patients presented with acute kidney injury (AKI) [KDIGO stage 3 in 31 (84%) patients] while neurological (n = 13, 36%) and cardiac (n = 7, 19%) damage were less frequent. ADAMTS13 (a disintegrin and metalloprotease with thrombospondin type I repeats-13) and complement activity were normal (n = 28 and 18 patients tested) and no pathogenic variant in the alternative complement pathway was found in 7 patients tested. TMA resolved in most (n = 34, 94%) patients but 12 (44%) still displayed stage 3 AKI at discharge. Nineteen (51%) patients were treated with therapeutic plasma exchange, 14 (38%) patients received corticosteroids and 18 (50%) were treated with eculizumab. However, none of these treatments demonstrated a significant impact on outcomes. CONCLUSION This study is the largest case series of CFZ-TMA since its approval in 2012. Patients present with severe AKI and experience frequent sequelae. Complement variants and blockade therapy do not seem to play a role in the pathophysiology and prognosis of the disease.
Collapse
Affiliation(s)
- Adrien Joseph
- Service de Médecine intensive réanimation, Hôpital Saint Louis, Assistance Publique des Hôpitaux de Paris, Paris, France
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Stéphanie Harel
- Service d'immuno-hématologie, Hôpital Saint Louis, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Laurent Mesnard
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), Assistance Publique des Hôpitaux de Paris, Paris, France
- Soins Intensifs Néphrologiques et Rein Aigu, Hôpital Tenon, Assistance Publique des Hôpitaux de Paris, Paris, France
- UMR_S1155, INSERM, Sorbonne Université, Paris, France
| | - Cédric Rafat
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), Assistance Publique des Hôpitaux de Paris, Paris, France
- Soins Intensifs Néphrologiques et Rein Aigu, Hôpital Tenon, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Silène Knapp
- Service d'immuno-hématologie, Hôpital Saint Louis, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Anne Rumpler
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), Assistance Publique des Hôpitaux de Paris, Paris, France
- Service d'hématologie, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | - Carole Philipponnet
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), Assistance Publique des Hôpitaux de Paris, Paris, France
- Service de Néphrologie, dialyse et transplantation, Centre Hospitalier Universitaire Clermont-Ferrand, Clermont-Ferrand, France
| | - Christophe Barba
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), Assistance Publique des Hôpitaux de Paris, Paris, France
- Service de Néphrologie et Nutrition, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Jean-Michel Rebibou
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), Assistance Publique des Hôpitaux de Paris, Paris, France
- Service de Néphrologie, Centre Hospitalier Universitaire Dijon Bourgogne, Dijon, France
| | - David Buob
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), Assistance Publique des Hôpitaux de Paris, Paris, France
- Service d'anatomo-pathologie, Hôpital Tenon, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Alexandre Hertig
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), Assistance Publique des Hôpitaux de Paris, Paris, France
- Service de néphrologie, Hôpital Foch, Suresnes, France
| | - Jacques Vargaftig
- Service d'hématologie, Institut Curie - Hôpital René Huguenin, Saint-Cloud, France
| | - Jean-Michel Halimi
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), Assistance Publique des Hôpitaux de Paris, Paris, France
- Service de Néphrologie-hypertension, Dialyses, Transplantation Rénale, Hôpital Bretonneau, Tours, France
- Hôpital Clocheville, Centre Hospitalier Universitaire de Tours, Tours, France et EA4245 T2i, Hôpital Trousseau, CHRU de Tours, Université de Tours, Tours, France
| | - Bertrand Arnulf
- Service d'immuno-hématologie, Hôpital Saint Louis, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Anne-Sophie Bretaud
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Bérangère Joly
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), Assistance Publique des Hôpitaux de Paris, Paris, France
- Service d'Hématologie Biologique, Hôpital Lariboisière, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Steven Grangé
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), Assistance Publique des Hôpitaux de Paris, Paris, France
- Service de Néphrologie, dialyse et transplantation, Centre Hospitalier Universitaire de Rouen, France
| | - Paul Coppo
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), Assistance Publique des Hôpitaux de Paris, Paris, France
- Service d'Hématologie, Hôpital Saint-Antoine, Assistance Publique des Hôpitaux de Paris, Paris, France
| |
Collapse
|
36
|
Yudaeva A, Kostyusheva A, Kachanov A, Brezgin S, Ponomareva N, Parodi A, Pokrovsky VS, Lukashev A, Chulanov V, Kostyushev D. Clinical and Translational Landscape of Viral Gene Therapies. Cells 2024; 13:1916. [PMID: 39594663 PMCID: PMC11592828 DOI: 10.3390/cells13221916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/29/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Gene therapies hold significant promise for treating previously incurable diseases. A number of gene therapies have already been approved for clinical use. Currently, gene therapies are mostly limited to the use of adeno-associated viruses and the herpes virus. Viral vectors, particularly those derived from human viruses, play a critical role in this therapeutic approach due to their ability to efficiently deliver genetic material to target cells. Despite their advantages, such as stable gene expression and efficient transduction, viral vectors face numerous limitations that hinder their broad application. These limitations include small cloning capacities, immune and inflammatory responses, and risks of insertional mutagenesis. This review explores the current landscape of viral vectors used in gene therapy, discussing the different types of DNA- and RNA-based viral vectors, their characteristics, limitations, and current medical and potential clinical applications. The review also highlights strategies to overcome existing challenges, including optimizing vector design, improving safety profiles, and enhancing transgene expression both using molecular techniques and nanotechnologies, as well as by approved drug formulations.
Collapse
Affiliation(s)
- Alexandra Yudaeva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
| | - Anastasiya Kostyusheva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
| | - Artyom Kachanov
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
| | - Sergey Brezgin
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (V.S.P.)
| | - Natalia Ponomareva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (V.S.P.)
- Department of Pharmaceutical and Toxicological Chemistry, Sechenov First Moscow State Medical University, 119146 Moscow, Russia
| | - Alessandro Parodi
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (V.S.P.)
| | - Vadim S. Pokrovsky
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (V.S.P.)
- Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
- Department of Biochemistry, People’s Friendship University, 117198 Moscow, Russia
| | - Alexander Lukashev
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
- Research Institute for Systems Biology and Medicine, 117246 Moscow, Russia
| | - Vladimir Chulanov
- Department of Infectious Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia;
| | - Dmitry Kostyushev
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (V.S.P.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
37
|
Germeni E, Cooper J, Briggs A, Laurence J. Treatment discontinuation in adults with atypical hemolytic uremic syndrome (aHUS): a qualitative study of international experts' perspectives with associated cost-consequence analysis. BMC Nephrol 2024; 25:411. [PMID: 39543505 PMCID: PMC11566521 DOI: 10.1186/s12882-024-03770-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 09/23/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Atypical hemolytic uremic syndrome (aHUS) is a rare, life-threatening thrombotic microangiopathy (TMA) related to congenital mutations impeding control of the alternative pathway of complement. Following approval of the complement C5 inhibitor eculizumab by the European Medicines Agency and the US Food and Drug Administration, initial guidelines suggested lifelong therapy. Yet, growing evidence indicates that discontinuation of eculizumab, or its long-acting form ravulizumab, is possible for many patients. This mixed-methods study sought to explore international experts' perspectives and experiences related to treatment duration in adult patients with aHUS, while also estimating the financial and potential health consequences of early discontinuation. METHODS Between January and December 2023, we conducted 10 qualitative interviews with experts in the treatment of aHUS, based upon which we constructed a quantitative decision tree, designed to estimate time on treatment and treatment- and disease-related adverse events. RESULTS Thematic analysis of the interview data identified four main themes: (1) Concerns and prior experience; (2) High-risk vs. low-risk groups; (3) Patient preference and adherence; and (4) Funding for monitoring and re-treatment. Although most interviewees were in favour of considering treatment discontinuation for many patients (citing the high cost, burden, and potential side effects of lifelong treatment as key reasons), a prior negative experience of discontinuation seemed to make others more reluctant to stop. Deciding which patients required lifelong treatment and which not involved consideration of a wide range of factors, including patient- and system-related factors. Cost-consequence analysis demonstrated the financial savings associated with early treatment discontinuation at the expense of increased risk of recurrent TMA events. Close monitoring for these events had the potential to minimise any long-term injury, primarily renal, with an estimated one event per 100 patient years. For patients at high risk of TMA and with poor adherence to monitoring, rates of renal injury rose to three events per 100 patient years. CONCLUSIONS aHUS treatment protocols are changing globally in response to new clinical evidence. Against this backdrop, our mixed-methods study provides compelling evidence on the complexity of factors influencing treatment discontinuation decisions in aHUS, as well as the financial and health consequences of early discontinuation.
Collapse
Affiliation(s)
- Evi Germeni
- Health Economics and Health Technology Assessment (HEHTA), School of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Jacie Cooper
- Avalon Health Economics LLC, Morristown, NJ, USA
| | - Andrew Briggs
- Department of Health Services Research and Policy, London School of Hygiene and Tropical Medicine, London, UK
| | - Jeffrey Laurence
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
38
|
Oakes A, Liu Y, Dubielecka PM. Complement or insult: the emerging link between complement cascade deficiencies and pathology of myeloid malignancies. J Leukoc Biol 2024; 116:966-984. [PMID: 38836653 PMCID: PMC11531810 DOI: 10.1093/jleuko/qiae130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
The complement cascade is an ancient and highly conserved arm of the immune system. The accumulating evidence highlights elevated activity of the complement cascade in cancer microenvironment and emphasizes its effects on the immune, cancer, and cancer stroma cells, pointing to a role in inflammation-mediated etiology of neoplasms. The role the cascade plays in development, progression, and relapse of solid tumors is increasingly recognized, however its role in hematological malignancies, especially those of myeloid origin, has not been thoroughly assessed and remains obscure. As the role of inflammation and autoimmunity in development of myeloid malignancies is becoming recognized, in this review we focus on summarizing the links that have been identified so far for complement cascade involvement in the pathobiology of myeloid malignancies. Complement deficiencies are primary immunodeficiencies that cause an array of clinical outcomes including an increased risk of a range of infectious as well as local or systemic inflammatory and thrombotic conditions. Here, we discuss the impact that deficiencies in complement cascade initiators, mid- and terminal-components and inhibitors have on the biology of myeloid neoplasms. The emergent conclusions indicate that the links between complement cascade, inflammatory signaling, and the homeostasis of hematopoietic system exist, and efforts should continue to detail the mechanistic involvement of complement cascade in the development and progression of myeloid cancers.
Collapse
Affiliation(s)
- Alissa Oakes
- Department of Medicine, Alpert Medical School, Brown University, 69 Brown St, Providence, RI 02906, USA
- Division of Hematology/Oncology, Rhode Island Hospital, 69 Brown St, Providence, RI 02906, USA
- Therapeutic Sciences Graduate program, Brown University, 69 Brown St, Providence, RI 02906, USA
| | - Yuchen Liu
- Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, 22. S. Greene St., Baltimore, MD 21201-1595, USA
| | - Patrycja M Dubielecka
- Department of Medicine, Alpert Medical School, Brown University, 69 Brown St, Providence, RI 02906, USA
- Division of Hematology/Oncology, Rhode Island Hospital, 69 Brown St, Providence, RI 02906, USA
- Therapeutic Sciences Graduate program, Brown University, 69 Brown St, Providence, RI 02906, USA
- Legorreta Cancer Center, Brown University, One Hoppin St., Coro West, Suite 5.01, Providence, RI 02903, USA
| |
Collapse
|
39
|
Maruyama S, Ikeda Y, Kaname S, Kato N, Matsumoto M, Ishikawa Y, Shimono A, Miyakawa Y, Nangaku M, Shibagaki Y, Okada H. Eculizumab for adult patients with atypical haemolytic-uraemic syndrome: full dataset analysis of Japanese post-marketing surveillance. J Nephrol 2024; 37:2181-2190. [PMID: 38809358 PMCID: PMC11649742 DOI: 10.1007/s40620-024-01921-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 02/10/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Eculizumab has been approved for atypical haemolytic-uraemic syndrome (aHUS) in Japan since 2013. Post-marketing surveillance enrolled patients with aHUS who received ≥ 1 dose of eculizumab to assess eculizumab safety and effectiveness. METHODS We evaluated serious adverse events and effectiveness endpoints, i.e., haematologic normalization, a decrease of ≥ 25% in serum creatinine (sCr) levels, and complete thrombotic microangiopathy (TMA) response in adult patients with aHUS without other underlying diseases. In addition, the difference of baseline characteristics between patients who did and did not meet effectiveness endpoints was examined. RESULTS In this safety and effectiveness analysis, 79 adult patients were included; median age was 54.0 years, median treatment duration was 30 weeks. Total exposure time of eculizumab was 75.51 patient-years, and 94 serious adverse events were reported in 39 patients. No unexpected safety signals were identified in this population. Mean platelet count, lactate dehydrogenase and estimated glomerular filtration rate significantly improved after 7 days of treatment. Complete TMA response, haematologic normalization and the improvement of sCr levels were met by 35.3%, 40.4% and 51.3% of patients, respectively. Median treatment duration was shorter in patients who did not achieve complete TMA response (6 weeks) than in patients who did (114 weeks). Multivariate analysis suggested that the time from the most recent TMA episode to start of eculizumab treatment was negatively associated with kidney function improvement. CONCLUSIONS No unexpected safety signals of eculizumab were identified in Japanese patients with aHUS in a real-world setting. Renal outcomes were negatively associated with the time from the most recent TMA episode to the initiation of eculizumab treatment.
Collapse
Affiliation(s)
- Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.
| | - Yoichiro Ikeda
- Division of Nephrology and Endocrinology, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Shinya Kaname
- Department of Nephrology and Rheumatology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-City, Tokyo, 181-8611, Japan
| | - Noritoshi Kato
- Department of Nephrology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Masanori Matsumoto
- Department of Blood Transfusion Medicine, Nara Medical University, 840 Shijyo-cho, Kashihara City, Nara, 634-8522, Japan
| | - Yumiko Ishikawa
- Alexion Pharma GK, 3-1-1 Shibaura, Minato-Ku, Tokyo, 108-0023, Japan
| | - Akihiko Shimono
- Alexion Pharma GK, 3-1-1 Shibaura, Minato-Ku, Tokyo, 108-0023, Japan
| | - Yoshitaka Miyakawa
- Department of Haematology, Saitama Medical University, 38 Moroyama, Iruma-gun, Saitama, 350-0495, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yugo Shibagaki
- Division of Nephrology and Hypertension, Department of Medicine, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Hirokazu Okada
- Department of Nephrology, Saitama Medical University, 38 Moroyama, Iruma-Gun, Saitama, 350-0495, Japan
| |
Collapse
|
40
|
Ferri M, Zotta F, Donadelli R, Dossier C, Duneton C, El-Sissy C, Fremeau-Bacchi V, Kwon T, Quadri L, Pasini A, Sellier-Leclerc AL, Vivarelli M, Hogan J. Anti-CFH-associated hemolytic uremic syndrome: do we still need plasma exchange? Pediatr Nephrol 2024; 39:3263-3269. [PMID: 38632123 DOI: 10.1007/s00467-024-06373-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Between 5 and 50% of atypical hemolytic uremic syndrome (aHUS) cases in children are caused by autoantibodies against complement factor H (CFH). Given the acquired autoimmune nature of the disease, plasma exchange (PE) and various immunosuppressive treatments have been used. More recently, eculizumab has been proposed. METHODS In this multicenter, retrospective study, we report outcomes of 12 children with anti-FH antibody-associated HUS treated with eculizumab associated with various immunosuppressive regimens. RESULTS Patients were treated with eculizumab for 15.5 [9.5;23.0] months and 3 received PE or IgG adsorption. Three patients received mycophenolate mofetil (MMF) alone, 1 patient received MMF and steroids, 1 patient received MMF and rituximab, 3 patients received MMF/steroids and rituximab, and 4 patients did not receive any immunosuppression. Anti-FH antibody levels significantly decreased but no difference was observed based on the immunosuppressive regimen. Eculizumab was discontinued in 7/10 patients after 11 [7.5;15.5] months and MMF in 6/8 patients after 36 [35;40] months. Anti-FH titers at MMF discontinuation ranged from 257 to 3425 UI/L. None of these patients relapsed and eGFR at last follow-up was above 70 mL/min/1.73 m2 in all patients. CONCLUSIONS Eculizumab is effective and safe in inducing and maintaining remission in aHUS secondary to anti-FH antibodies and renders reduction of anti-FH titers less urgent. Anti-FH antibody titers decreased in most patients irrespective of the immunosuppressive treatment chosen, so that a strategy consisting of combining eculizumab with MMF monotherapy seems sufficient at least in non-Indian or less severe forms of anti-FH antibody-associated HUS.
Collapse
Affiliation(s)
- Marion Ferri
- Pediatric Nephrology Department, Centre de référence des Maladies Rénales Rares MARHEA, Filières Maladies Rares ORKID et ERK-Net, Robert Debré Hospital, APHP, 48 bd Sérurier, 75019, Paris, France
| | - Federica Zotta
- Division of Nephrology, Laboratory of Nephrology, Ospedale Pediatrico Bambino Gesù IRCCS, Rome, Italy
| | - Roberta Donadelli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Ranica, Bergamo, Italy
| | - Claire Dossier
- Pediatric Nephrology Department, Centre de référence des Maladies Rénales Rares MARHEA, Filières Maladies Rares ORKID et ERK-Net, Robert Debré Hospital, APHP, 48 bd Sérurier, 75019, Paris, France
| | - Charlotte Duneton
- Pediatric Nephrology Department, Centre de référence des Maladies Rénales Rares MARHEA, Filières Maladies Rares ORKID et ERK-Net, Robert Debré Hospital, APHP, 48 bd Sérurier, 75019, Paris, France
| | - Carine El-Sissy
- Assistance Publique-Hôpitaux de Paris, Department of Biological Immunology, Hôpital Européen Georges Pompidou, and INSERM UMR S1138, Complément et Maladies, Centre de Recherche des Cordeliers, Paris, France
| | - Véronique Fremeau-Bacchi
- Assistance Publique-Hôpitaux de Paris, Department of Biological Immunology, Hôpital Européen Georges Pompidou, and INSERM UMR S1138, Complément et Maladies, Centre de Recherche des Cordeliers, Paris, France
| | - Thérésa Kwon
- Pediatric Nephrology Department, Centre de référence des Maladies Rénales Rares MARHEA, Filières Maladies Rares ORKID et ERK-Net, Robert Debré Hospital, APHP, 48 bd Sérurier, 75019, Paris, France
| | - Lisa Quadri
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Ranica, Bergamo, Italy
| | - Andrea Pasini
- Paediatric Clinic, IRCCS Ospedale Maggiore Policlinico Sant'Orsola, Department of Medicine and Surgery, University of Bologna, Bologna, Italy
| | - Anne-Laure Sellier-Leclerc
- Department of Pediatric Nephrology, Centre de Référence des Maladies Rénales Rares Néphrogones, Filières Maladies Rares ORKID et ERK-Net, CHU de Lyon, Bron, France
| | - Marina Vivarelli
- Division of Nephrology, Laboratory of Nephrology, Ospedale Pediatrico Bambino Gesù IRCCS, Rome, Italy
| | - Julien Hogan
- Pediatric Nephrology Department, Centre de référence des Maladies Rénales Rares MARHEA, Filières Maladies Rares ORKID et ERK-Net, Robert Debré Hospital, APHP, 48 bd Sérurier, 75019, Paris, France.
- INSERM, UMR-U970, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université Paris Cité, Paris, France.
| |
Collapse
|
41
|
Azoulay E, Zuber J, Bousfiha AA, Long Y, Tan Y, Luo S, Essafti M, Annane D. Complement system activation: bridging physiology, pathophysiology, and therapy. Intensive Care Med 2024; 50:1791-1803. [PMID: 39254734 DOI: 10.1007/s00134-024-07611-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/10/2024] [Indexed: 09/11/2024]
Abstract
The complement system is a set of over 50 proteins that constitutes an essential part of the innate immune system. Complement system activation involves an organized proteolytic cascade. Overactivation of complement system activation is the main pathogenic mechanism of several diseases and contributes to the manifestations of many other conditions. This review describes the normal complement system and the role for complement dysregulation in critical illnesses, notably sepsis and acute respiratory distress syndrome. Complement activation is involved in the immune system response to pathogens but, when excessive, can contribute to tissue damage, runaway inflammation, and capillary leakage syndrome. Complement overactivation may play a key role in severe forms of coronavirus disease 2019 (COVID-19). Two diseases whose manifestations are mainly caused by complement overactivation, namely, atypical hemolytic and uremic syndrome (aHUS) and myasthenia gravis, are discussed. A diagnostic algorithm for aHUS is provided. Early complement-inhibiting therapy has been proven effective. When renal transplantation is required, complement-inhibiting drugs can be used prophylactically to prevent aHUS recurrence. Similarly, acetylcholine-receptor autoantibody-positive generalized myasthenia gravis involves complement system overactivation and responds to complement inhibition. The two main complement inhibitors used in to date routine are eculizumab and ravulizumab. The main adverse event is Neisseria infection, which is rare and preventable, but can be fatal. The complement system is crucial to health but, when overactivated, can cause or contribute to disease. Effective complement inhibitors are now available, although additional data are required to determine optimal regimens. Further research is also needed to better understand the complement system, develop advanced diagnostic tools, and identify markers that allow the personalization of treatment strategies.
Collapse
Affiliation(s)
- Elie Azoulay
- Intensive Care Unit, Saint-Louis University Hospital, AP-HP, Paris Cité University, Paris, France.
| | - Julien Zuber
- Department of Kidney and Metabolic Diseases, Transplantation and Clinical Immunology, Necker University Hospital, AP-HP, Paris, France
| | - Ahmed Aziz Bousfiha
- Department of Pediatric Infectious and Immunological Diseases, IbnRochd University Hospital, Casablanca, Morocco
- Laboratory of Clinical Immunology, Inflammation and Allergy (LICIA), Casablanca, Morocco
- School of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Yun Long
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Tan
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, PR China
- Institute of Nephrology, Peking University, Beijing, PR China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, PR China
| | - Sushan Luo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, PR China
- Huashan Rare Diseases Center, Huashan Hospital, Fudan University, Shanghai, PR China
- National Center for Neurological Diseases, Shanghai, PR China
| | - Meriem Essafti
- Intensive Care Department, Mother-Children Center, Mohamed VI University Hospital, Marrakech, Morocco
| | - Djillali Annane
- Department of Intensive Care, Raymond Poincaré Hospital, AP-HP, Garches, France
- Simone Veil School of Medicine, Versailles-Saint Quentin University, Paris-Saclay University, Versaillles, France
- Institut Hospitalo-Universitaire PROMETHEUS & Fédération Hospitalo-Universitaire SEPSIS, Paris-Saclay University, Saclay, France
- INSERM, Garches, France
| |
Collapse
|
42
|
Rashid S, Ahmed S, Ahmed Khan M. A Case of Thrombotic Microangiopathy Secondary to Hypertensive Emergency: Presentation, Management, and Distinguishing Features. Cureus 2024; 16:e74498. [PMID: 39726463 PMCID: PMC11670735 DOI: 10.7759/cureus.74498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Thrombotic microangiopathies (TMA) are a group of conditions that present with varying degrees of microthrombi, thrombocytopenia, microangiopathic hemolytic anemia, renal dysfunction, and neurological impairment. Etiologies can be primary, such as thrombotic thrombocytopenic purpura (TTP), hemolytic uremic syndrome (HUS), and atypical hemolytic uremic syndrome (aHUS), or secondary, such as due to systemic infections, malignancies, immune-mediated conditions, and hypertensive emergencies. In hypertensive emergencies, this presentation can occur from mechanical stress placed on red blood cells as they pass through narrowed arteries due to edema and microangiopathic changes within the vessels themselves. In TMA secondary to hypertensive emergencies (HTN-TMA), blood pressure control alone can lead to improvement in cytopenias. We present a case of a 48-year-old male with HTN-TMA. The patient had normalization of thrombocytopenia and improvement in anemia with adequate blood pressure control. This case highlights the difficulty in making this diagnosis due to overlapping presentations with primary thrombotic microangiopathies and the extensive etiologies that should be considered as part of a differential diagnosis.
Collapse
Affiliation(s)
- Saad Rashid
- Department of Internal Medicine, Mercyhealth Graduate Medical Education (GME) Consortium, Rockford, USA
| | - Sultan Ahmed
- Department of Internal Medicine, Mercyhealth Graduate Medical Education (GME) Consortium, Rockford, USA
- College of Medicine, Allegheny Health Network, Erie, USA
- College of Medicine, Lake Erie College of Osteopathic Medicine, Erie, USA
| | - Mohammed Ahmed Khan
- Department of Medicine, Mercyhealth Graduate Medical Education (GME) Consortium, Rockford, USA
| |
Collapse
|
43
|
Heidenreich K, Goel D, Priyamvada PS, Kulkarni S, Chakurkar V, Khullar D, Singh R, Bale C, Zipfel PF. C3 glomerulopathy: a kidney disease mediated by alternative pathway deregulation. FRONTIERS IN NEPHROLOGY 2024; 4:1460146. [PMID: 39534179 PMCID: PMC11554616 DOI: 10.3389/fneph.2024.1460146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/17/2024] [Indexed: 11/16/2024]
Abstract
C3 glomerulopathy (C3G) is an ultra-rare complement-mediated kidney disease caused by to the deregulation of the alternative pathway (AP) of proximal complement. Consequently, all effector loops of the complement are active and can lead to pathologies, such as C3a- and C5a-mediated inflammation, C3b opsonization, surface C3b-mediated AP C3 convertase assembly, C3 cleavage product deposition in the glomerulus, and lytic C5b-9/MAC cell damage. The most common pathologic mechanisms are defective chronic alternative pathway deregulation, mostly occurring in the plasma, often causing C3 consumption, and chronic complement-mediated glomerular damage. C3G develops over several years, and loss of renal function occurs in more than 50% of patients. C3G is triggered by both genetic and autoimmune alterations. Genetic causes include mutations in individual complement genes and chromosomal variations in the form of deletions and duplications affecting genes encoding complement modulators. Many genetic aberrations result in increased AP C3 convertase activity, either due to decreased activity of regulators, increased activity of modulators, or gain-of-function mutations in genes encoding components of the convertase. Autoimmune forms of C3G do also exist. Autoantibodies target individual complement components and regulators or bind to neoepitopes exposed in the central alternative pathway C3 convertase, thereby increasing enzyme activity. Overactive AP C3 convertase is common in C3G patients. Given that C3G is a complement disease mediated by defective alternative pathway action, complement blockade is an emerging concept for therapy. Here, we summarize both the causes of C3G and the rationale for complement inhibition and list the inhibitors that are being used in the most advanced clinical trials for C3G. With several inhibitors in phase II and III trials, it is expected that effectice treatment for C3G will become availabe in the near future.
Collapse
Affiliation(s)
| | | | - P. S. Priyamvada
- Department of Nephrology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Sagar Kulkarni
- Department of Nephrology, King Edward Memorial Hospital, Pune, Maharashtra, India
| | - Vipul Chakurkar
- Department of Nephrology, King Edward Memorial Hospital, Pune, Maharashtra, India
| | - Dinesh Khullar
- Department of Nephrology and Renal Transplantation, Max Super Speciality Hospital Saket, New Delhi, India
| | - Ravi Singh
- Department of Nephrology and Renal Transplant, Jaypee Hospital, Noida, Uttar Pradesh, India
| | - Charan Bale
- Department of Nephrology, Dr. D.Y. Patil Medical College & Research Centre, Pune, Maharashtra, India
| | - Peter F. Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| |
Collapse
|
44
|
Matsumoto M, Shimono A, Yokosawa J, Hirose K, Wang E, Maruyama S. Correlation between a 2-week change in platelet count and clinical outcomes after the initiation of ravulizumab treatment in adult patients with atypical hemolytic uremic syndrome: post-hoc analysis of the phase III trial. Thromb J 2024; 22:93. [PMID: 39468592 PMCID: PMC11520077 DOI: 10.1186/s12959-024-00652-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/09/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Atypical hemolytic uremic syndrome (aHUS) is a rare disease with poor outcomes when untreated, in which ravulizumab or eculizumab are the standard of care where available. It has been proposed to regularly monitor platelet counts as an early response to ravulizumab or eculizumab. This study aimed to investigate the association between the early response to ravulizumab treatment and renal outcomes through 26 weeks in complement inhibitor-naïve adults with aHUS. METHODS Adult patients with aHUS enrolled in the ALXN1210-aHUS-311 phase III study of ravulizumab were divided into two groups according to the achievement of complete thrombotic microangiopathy (TMA) response, i.e., platelet count and lactate dehydrogenase (LDH) normalization and ≥ 25% improvement in serum creatinine (sCr) from baseline, by 26 weeks and baseline characteristics were compared. Changes in hematologic parameters, platelet count and LDH, were compared between the two groups. Finally, we examined whether early hematologic improvement was associated with renal recovery (dialysis discontinuation or ≥ 25% improvement in sCr from baseline) through 26 weeks. RESULTS Of 56 ravulizumab-treated patients, 30 achieved complete TMA response for 26 weeks, and 26 did not. Patients with complete TMA response showed rapid improvements in platelet counts. In patients without complete TMA response, delayed normalization of platelet counts was observed. By day 15, 93.3% (28/30) of patients with complete TMA response at 26 weeks and 26.9% (7/26) of patients without complete TMA response achieved platelet normalization. At 26 weeks, 62.5% (35/56) achieved renal recovery; however, 37.5% (21/56) did not. In patients with renal recovery, 85.7% (30/35) of patients had platelet count normalization by day 15; in patients without renal recovery, 23.8% (5/21) of patients had platelet count normalization (P < 0.0001). Receiver operator characteristic curve analysis showed a moderate association between platelet counts on day 8/15 and renal recovery within 26 weeks (day 8: area under the curve [AUC] = 0.7985; day 15: AUC = 0.8406). CONCLUSIONS Platelet count normalization occurred in 62.5% (35/56) by day 15 after ravulizumab initiation and was associated with renal recovery through 26 weeks in complement inhibitor-naïve adults with aHUS. TRIAL REGISTRATION This study was performed as a post-hoc analysis of the ALXN1210-aHUS-311 phase III clinical trial (NCT02949128, registered October 25, 2016).
Collapse
Affiliation(s)
- Masanori Matsumoto
- Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8521, Japan.
| | - Akihiko Shimono
- Alexion Pharma GK, 3-1-1 Shibaura, Minato-ku, Tokyo, 108-0023, Japan
| | - Jun Yokosawa
- Alexion Pharma GK, 3-1-1 Shibaura, Minato-ku, Tokyo, 108-0023, Japan
| | - Keiichiro Hirose
- Alexion Pharma GK, 3-1-1 Shibaura, Minato-ku, Tokyo, 108-0023, Japan
| | - Edward Wang
- Alexion, AstraZeneca Rare Disease, 121 Seaport Blvd, Boston, MA 02210, USA
| | - Shoichi Maruyama
- Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
45
|
Meliambro K, He JC, Campbell KN. Podocyte-targeted therapies - progress and future directions. Nat Rev Nephrol 2024; 20:643-658. [PMID: 38724717 DOI: 10.1038/s41581-024-00843-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2024] [Indexed: 09/14/2024]
Abstract
Podocytes are the key target cells for injury across the spectrum of primary and secondary proteinuric kidney disorders, which account for up to 90% of cases of kidney failure worldwide. Seminal experimental and clinical studies have established a causative link between podocyte depletion and the magnitude of proteinuria in progressive glomerular disease. However, no substantial advances have been made in glomerular disease therapies, and the standard of care for podocytopathies relies on repurposed immunosuppressive drugs. The past two decades have seen a remarkable expansion in understanding of the mechanistic basis of podocyte injury, with prospects increasing for precision-based treatment approaches. Dozens of disease-causing genes with roles in the pathogenesis of clinical podocytopathies have been identified, as well as a number of putative glomerular permeability factors. These achievements, together with the identification of novel targets of podocyte injury, the development of potential approaches to harness the endogenous podocyte regenerative potential of progenitor cell populations, ongoing clinical trials of podocyte-specific pharmacological agents and the development of podocyte-directed drug delivery systems, contribute to an optimistic outlook for the future of glomerular disease therapy.
Collapse
Affiliation(s)
- Kristin Meliambro
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John C He
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kirk N Campbell
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
46
|
Smith J, Hans V, Yacyshyn E, Rouhi A, Oliver M. Systemic lupus erythematosus presenting with atypical hemolytic uremic syndrome: a case report and review of the literature. Rheumatol Int 2024; 44:2213-2225. [PMID: 38502235 DOI: 10.1007/s00296-024-05558-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 02/09/2024] [Indexed: 03/21/2024]
Abstract
Systemic lupus erythematosus (SLE) can present with a diverse array of hematologic manifestations, among which atypical hemolytic uremic syndrome (aHUS) is a rare entity. SLE-triggered aHUS has significant morbidity and mortality without timely intervention, yet its frequency remains uncertain and optimal strategies for complement-directed therapies are largely expert-driven. We performed a comprehensive literature review and present a case of a 23-year-old female newly diagnosed with SLE/class IV lupus nephritis who developed aHUS that rapidly responded to the C5 antagonist, eculizumab. Review of the current literature identified forty-nine published cases of SLE with concurrent aHUS and revealed a predilection for aHUS in younger SLE patients, concurrent presentation with lupus nephritis, anti-dsDNA positivity, and complement system abnormalities. Over seventy percent of cases used eculizumab as complement-directed therapy with a trend towards faster time to improvement in laboratory parameters, though reported outcomes were highly variable. Early recognition of aHUS in SLE is pivotal in guiding appropriate therapeutic interventions, and prompt initiation of eculizumab may reduce the potential morbidity associated with plasmapheresis and additional immunosuppression. While eculizumab showcases promising results, its optimal timing and duration remain elusive. An understanding of a patients' complement genetics could aid management strategies, and ongoing research into complement-targeted therapies offers promising avenues for both SLE and aHUS treatment.
Collapse
Affiliation(s)
- Justin Smith
- Department of Medicine, University of Alberta, 8-130 Clinical Sciences Building, 11350 83 Avenue NW, Edmonton, AB, T6G 2G3, Canada.
| | - Varinder Hans
- Department of Medicine, University of Alberta, 8-130 Clinical Sciences Building, 11350 83 Avenue NW, Edmonton, AB, T6G 2G3, Canada
| | - Elaine Yacyshyn
- Department of Medicine, University of Alberta, 8-130 Clinical Sciences Building, 11350 83 Avenue NW, Edmonton, AB, T6G 2G3, Canada
| | - Azin Rouhi
- Department of Medicine, University of Alberta, 8-130 Clinical Sciences Building, 11350 83 Avenue NW, Edmonton, AB, T6G 2G3, Canada
| | - Monika Oliver
- Department of Medicine, Division of Hematology, University of Alberta, Edmonton, Canada
| |
Collapse
|
47
|
Celegen K, Gulhan B, Fidan K, Yuksel S, Yilmaz N, Yılmaz AC, Demircioğlu Kılıç B, Gokce I, Kavaz Tufan A, Kalyoncu M, Nalcacıoglu H, Ozlu SG, Kurt Sukur ED, Canpolat N, K Bayazit A, Çomak E, Tabel Y, Tulpar S, Celakil M, Bek K, Zeybek C, Duzova A, Özçakar ZB, Topaloglu R, Soylemezoglu O, Ozaltin F. Adolescence-onset atypical hemolytic uremic syndrome: is it different from infant-onset? Clin Exp Nephrol 2024; 28:1027-1037. [PMID: 38704765 DOI: 10.1007/s10157-024-02505-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 04/08/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND Atypical hemolytic uremic syndrome (aHUS) is a rare, mostly complement-mediated thrombotic microangiopathy. The majority of patients are infants. In contrast to infantile-onset aHUS, the clinical and genetic characteristics of adolescence-onset aHUS have not been sufficiently addressed to date. METHODS A total of 28 patients (21 girls, 7 boys) who were diagnosed as aHUS between the ages of ≥10 years and <18 years were included in this study. All available data in the Turkish Pediatric aHUS registry were collected and analyzed. RESULTS The mean age at diagnosis was 12.8±2.3 years. Extra-renal involvement was noted in 13 patients (46.4%); neurological involvement was the most common (32%). A total of 21 patients (75%) required kidney replacement therapy. Five patients (17.8%) received only plasma therapy and 23 (82%) of the patients received eculizumab. Hematologic remission and renal remission were achieved in 25 (89.3%) and 17 (60.7%) of the patients, respectively. Compared with the infantile-onset aHUS patients, adolescent patients had a lower complete remission rate during the first episode (p = 0.002). Genetic analyses were performed in all and a genetic variant was detected in 39.3% of the patients. The mean follow-up duration was 4.9±2.6 years. At the last visit, adolescent patients had lower eGFR levels (p = 0.03) and higher rates of chronic kidney disease stage 5 when compared to infantile-onset aHUS patients (p = 0.04). CONCLUSIONS Adolescence-onset aHUS is a rare disease but tends to cause more permanent renal dysfunction than infantile-onset aHUS. These results may modify the management approaches in these patients.
Collapse
Affiliation(s)
- Kubra Celegen
- Department of Pediatric Nephrology, Kayseri Education and Research Hospital, Kayseri, Türkiye
| | - Bora Gulhan
- Department of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, 06100, Sihhiye, Ankara, Turkey.
| | - Kibriya Fidan
- Department of Pediatric Nephrology, Faculty of Medicine, Gazi University, Ankara, Türkiye
| | - Selcuk Yuksel
- Department of Pediatric Nephrology, Faculty of Medicine, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| | - Neslihan Yilmaz
- Department of Pediatric Nephrology, Necip Fazil City Hospital, Kahramanmaras, Türkiye
| | - Aysun Caltik Yılmaz
- Department of Pediatric Nephrology, Faculty of Medicine, Baskent University, Ankara, Türkiye
| | | | - Ibrahim Gokce
- Department of Pediatric Nephrology, Faculty of Medicine, Marmara University, Istanbul, Türkiye
| | - Aslı Kavaz Tufan
- Department of Pediatric Nephrology, Faculty of Medicine, Osmangazi University, Eskisehir, Türkiye
| | - Mukaddes Kalyoncu
- Department of Pediatric Nephrology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Türkiye
| | - Hulya Nalcacıoglu
- Department of Pediatric Nephrology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Türkiye
| | - Sare Gulfem Ozlu
- Department of Pediatric Nephrology, Ankara City Training and Research Hospital, Ankara, Türkiye
| | - Eda Didem Kurt Sukur
- Department of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Nur Canpolat
- Department of Pediatric Nephrology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, İstanbul, Türkiye
| | - Aysun K Bayazit
- Department of Pediatric Nephrology, Faculty of Medicine, Cukurova University, Adana, Türkiye
| | - Elif Çomak
- Department of Pediatric Nephrology, Faculty of Medicine, Akdeniz University, Antalya, Türkiye
| | - Yılmaz Tabel
- Department of Pediatric Nephrology, Faculty of Medicine, Inonu University, Malatya, Türkiye
| | - Sebahat Tulpar
- Department of Pediatric Nephrology, Istanbul Bakirkoy Dr. Sadi Konuk Training and Research Hospital, University of Health Sciences, İstanbul, Türkiye
| | - Mehtap Celakil
- Department of Pediatric Nephrology, Sakarya University Training and Research Hospital, Sakarya, Türkiye
| | - Kenan Bek
- Department of Pediatric Nephrology, Faculty of Medicine, Kocaeli University, Kocaeli, Türkiye
| | - Cengiz Zeybek
- Department of Pediatric Nephrology, Gulhane Training and Research Hospital, Ankara, Türkiye
| | - Ali Duzova
- Department of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Zeynep Birsin Özçakar
- Department of Pediatric Nephrology, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | - Rezan Topaloglu
- Department of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Oguz Soylemezoglu
- Department of Pediatric Nephrology, Faculty of Medicine, Gazi University, Ankara, Türkiye
| | - Fatih Ozaltin
- Department of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
- Nephrogenetics Laboratory, Faculty of Medicine, Hacettepe University, Ankara, Türkiye
- Center for Genomics and Rare Diseases, Hacettepe University, Ankara, Türkiye
- Department of Bioinformatics, Hacettepe University Institute of Health Sciences, Ankara, Türkiye
| |
Collapse
|
48
|
Jiang L, Wang S, Tan Y, Su T. Postpartum Renal Cortical Necrosis: A Case Series. Kidney Med 2024; 6:100892. [PMID: 39314861 PMCID: PMC11417324 DOI: 10.1016/j.xkme.2024.100892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024] Open
Abstract
Rationale & Objective Postpartum renal cortical necrosis (postpartum RCN) is a severe form of obstetric acute kidney injury. This study aimed to identify clinicopathologic features in Chinese postpartum RCN cases to determine how pathologic findings may contribute to the treatment and prognosis. Study Design Single-center, case series. Setting & Participants Twelve patients with postpartum RCN had kidney biopsies at Peking University First Hospital between 2014 and 2021. The diagnosis of postpartum RCN was made according to typical magnetic resonance imaging or pathologic features. Clinical, laboratory, and pathologic data were compared between patients with estimated glomerular filtration rate <30 (poor outcome) and ≥30 mL/min/1.73 m2 after 6 months. Observations All patients with postpartum RCN presented with stage 3 acute kidney injury attributed to a probable atypical hemolytic uremic syndrome. Pregnancy terminations occurred at a median gestational age of 35.5 weeks. Kidney biopsy was performed from 18 days to 4 months from delivery. On biopsy, hemoglobin, platelet count, and lactate dehydrogenase levels had been restored to 137 g/L, 214 × 109/L, and 231.50 ± 65.01 U/L, respectively. Four patients exhibited poor outcome, demonstrating higher schistocyte count, serum creatinine, and mean arterial pressure at onset. Pathologically, glomerular segmental sclerosis was prevalent. The "not otherwise specified" variant was the most common type, followed by collapsing variant, cellular variant, and tip variant. Patients with poor kidney outcome had more glomerular coagulative necrosis, capillary thrombosis, extensive cortical coagulative necrosis, and pronounced arteriole/artery lesions including increased interlobular arteriole intimal edema and fibrin thrombosis, but a lower occurrence of segmental sclerosis. Limitations Limited sample size and retrospective design. Conclusions We identified key pathologic features in patients with postpartum RCN and atypical hemolytic uremic syndrome, highlighting the necessity for more effective therapeutic options. There is a clear demand for noninvasive biomarkers that can accurately track disease progression and inform treatment duration for long-term outcomes improvement.
Collapse
Affiliation(s)
- Lei Jiang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Renal Pathology Center, Institute of Nephrology, Peking University, Beijing, China
| | - Suxia Wang
- Renal Pathology Center, Institute of Nephrology, Peking University, Beijing, China
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, China
| | - Ying Tan
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Renal Pathology Center, Institute of Nephrology, Peking University, Beijing, China
| | - Tao Su
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Renal Pathology Center, Institute of Nephrology, Peking University, Beijing, China
| |
Collapse
|
49
|
Tsakiris DA, Gavriilaki E, Chanou I, Meyer SC. Hemostasis and complement in allogeneic hematopoietic stem cell transplantation: clinical significance of two interactive systems. Bone Marrow Transplant 2024; 59:1349-1359. [PMID: 39004655 PMCID: PMC11452340 DOI: 10.1038/s41409-024-02362-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024]
Abstract
Hematopoietic stem cell transplantation (HCT) represents a curative treatment option for certain malignant and nonmalignant hematological diseases. Conditioning regimens before HCT, the development of graft-versus-host disease (GVHD) in the allogeneic setting, and delayed immune reconstitution contribute to early and late complications by inducing tissue damage or humoral alterations. Hemostasis and/or the complement system are biological regulatory defense systems involving humoral and cellular reactions and are variably involved in these complications after allogeneic HCT. The hemostasis and complement systems have multiple interactions, which have been described both under physiological and pathological conditions. They share common tissue targets, such as the endothelium, which suggests interactions in the pathogenesis of several serious complications in the early or late phase after HCT. Complications in which both systems interfere with each other and thus contribute to disease pathogenesis include transplant-associated thrombotic microangiopathy (HSCT-TMA), sinusoidal obstruction syndrome/veno-occlusive disease (SOS/VOD), and GVHD. Here, we review the current knowledge on changes in hemostasis and complement after allogeneic HCT and how these changes may define clinical impact.
Collapse
Affiliation(s)
| | - Eleni Gavriilaki
- Second Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioanna Chanou
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, Thessaloniki, Greece
| | - Sara C Meyer
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
50
|
Uriol-Rivera MG, Andrade BL, Bonet AM, Mulet AO, Ruiz CB, Parraga LP, Lumbreras J, Rota JIA, Servalos MF, Balaguer JF, Ferreres LP, Valles MJP, Valero RMRDG, Sanchez ST, Martin AG, Garcia JR, Cobo CG, Ramis-Cabrer D. Risk factors of death or chronic renal replacement therapy requirements in patients with thrombotic microangiopathies without ADAMTS-13 deficiency. Eur J Haematol 2024; 113:510-520. [PMID: 38955806 DOI: 10.1111/ejh.14261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 07/04/2024]
Abstract
Thrombotic microangiopathy (TMA), characterized by microangiopathic hemolytic anemia, thrombocytopenia, and multisystem organ dysfunction, is a life-threatening disease. Patients with TMA who do not exhibit a severe ADAMTS-13 deficiency (defined as a disintegrin-like and metalloprotease with thrombospondin type 1 motif no. 13 activity ≥10%: TMA-13n) continue to experience elevated mortality rates. This study explores the prognostic indicators for augmented mortality risk or necessitating chronic renal replacement therapy (composite outcome: CO) in TMA-13n patients. We included 42 TMA-13n patients from January 2008 to May 2018. Median age of 41 years and 60% were female. At presentation, 62% required dialysis, and 57% warranted intensive care unit admission. CO was observed in 45% of patients, including a 9-patient mortality subset. Multivariate logistic regression revealed three independent prognostic factors for CO: early administration of eculizumab (median time from hospitalization to eculizumab initiation: 5 days, range 0-19 days; odds ratio [OR], 0.14; 95% confidence interval [CI], 0.02-0.94), presence of neuroradiological lesions (OR, 6.67; 95% CI, 1.12-39.80), and a PLASMIC score ≤4 (OR, 7.39; 95% CI, 1.18-46.11). In conclusion, TMA-13n patients exhibit a heightened risk of CO in the presence of low PLASMIC scores and neuroradiological lesions, while early eculizumab therapy was the only protective factor.
Collapse
Affiliation(s)
- Miguel G Uriol-Rivera
- Nephrology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Bernardo López Andrade
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
- Hematology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Antonio Mas Bonet
- Radiology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Aina Obrador Mulet
- Nephrology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Carmen Ballester Ruiz
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
- Hematology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Leonor Periañez Parraga
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
- Pharmacy Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Javier Lumbreras
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
- Pediatric Nephrology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - José Ignacio Ayestarán Rota
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
- Intensive Care Unit, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | | | - Joana Ferrer Balaguer
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
- Immunology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Lucio Pallares Ferreres
- Internal Medicine Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - María Jose Picado Valles
- Radiology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | | | - Susana Tarongi Sanchez
- Neurology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Ana Garcia Martin
- Neurology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Juan Rodríguez Garcia
- Preventive Medicine Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Cristina Gomez Cobo
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
- Clinical Analysis, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Daniel Ramis-Cabrer
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| |
Collapse
|