1
|
Li X, Zhu L. Comprehensive profiling of cell type-specific expression and distribution of complement genes in mouse and human kidneys: insights into normal physiology and response to kidney transplantations. Ren Fail 2025; 47:2471568. [PMID: 40015727 PMCID: PMC11869339 DOI: 10.1080/0886022x.2025.2471568] [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: 07/24/2024] [Revised: 01/24/2025] [Accepted: 02/15/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Recent studies innovatively revealed the localized expression of complement genes in kidneys and shed light on the vital roles of the intracellular complement system in the physiologic function and pathological conditions. However, a comprehensive analysis of the expression of complement genes in the context of the evolving cellular landscape of the kidney is not available. METHODS We analyzed single-cell RNA sequencing data from healthy human subjects, C57BL/6 mice, and kidney transplant-rejected mice. The data were sourced from the NCBI Gene Expression Omnibus and processed using quality control measures and unsupervised clustering. Differential gene analyses were based on expression levels. RESULTS In total, 50 complement genes were categorized into pattern recognition molecules, proteases, complement components, receptors, and regulators. In normal mice kidneys, complement genes were expressed at relatively low levels. Among different complement gene categories, receptor genes were most widely expressed in kidney cells. Comparatively, macrophages and mesangial cells are the most abundant immune and nonimmune cell types for complement gene expression. A comparison of human and mouse data showed similar expression patterns, but human kidney complement gene expression was more abundant. Comparative analysis between mouse transplant-rejected and normal kidneys demonstrated stronger complement gene expression in transplant-rejected kidneys. CONCLUSIONS This study illustrated significant similarities in complement gene expression between murine and human kidneys and highlighted the responsive nature of complement genes to kidney injury, underscoring the dynamic nature of local complement regulation. These findings enhance our understanding of the complex regulation of the complement system within the kidney, offering insights into its role in renal disease pathogenesis.
Collapse
Affiliation(s)
- Xianzhi Li
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Li Zhu
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| |
Collapse
|
2
|
Welsh SJ, Zhang Y, Smith RJH. Acquired drivers of C3 glomerulopathy. Clin Kidney J 2025; 18:sfaf022. [PMID: 40052168 PMCID: PMC11883229 DOI: 10.1093/ckj/sfaf022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Indexed: 03/09/2025] Open
Abstract
C3 glomerulopathy (C3G) is a group of heterogeneous ultrarare kidney diseases characterized by dysregulated activation of the complement alternative pathway (AP) leading to excessive C3 cleavage. Diagnosis relies on kidney biopsy showing predominant C3 deposition in the glomerular basement membrane, with electron microscopy differentiating between dense deposit disease (DDD) and C3 glomerulonephritis (C3GN). The main drivers of AP dysregulation in C3G are acquired rather than genetic and consist primarily of autoantibodies called nephritic factors (C3Nefs, C4Nefs and C5Nefs) that bind to and stabilize complement convertases, causing complement overactivation. Current therapies are largely supportive, and existing complement-targeting treatments, such as eculizumab, demonstrate limited efficacy. Challenges in studying C3G include variability in autoantibody detection and a lack of standardized assays, which complicates clinical interpretation. Comprehensive assessment involving autoantibody panels, complement biomarkers, functional assays and genetic testing provides a more complete understanding of disease dynamics; however, key knowledge gaps remain regarding Nef origins, mechanisms and their pathogenic role. In this review we discuss acquired drivers of C3G with an emphasis on C3Nefs and C5Nefs and suggest areas of interest that might benefit from future research.
Collapse
Affiliation(s)
- Seth J Welsh
- Department of Internal Medicine, Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Yuzhou Zhang
- Department of Internal Medicine, Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Richard J H Smith
- Department of Internal Medicine, Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
3
|
Ricklin D. Complement-targeted therapeutics: Are we there yet, or just getting started? Eur J Immunol 2024; 54:e2350816. [PMID: 39263829 PMCID: PMC11628912 DOI: 10.1002/eji.202350816] [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: 06/03/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
Therapeutic interventions in the complement system, a key immune-inflammatory mediator and contributor to a broad range of clinical conditions, have long been considered important yet challenging or even unfeasible to achieve. Almost 20 years ago, a spark was lit demonstrating the clinical and commercial viability of complement-targeted therapies. Since then, the field has experienced an impressive expansion of targeted indications and available treatment modalities. Currently, a dozen distinct complement-specific therapeutics covering several intervention points are available in the clinic, benefiting patients suffering from eight disorders, not counting numerous clinical trials and off-label uses. Observing this rapid rise of complement-targeted therapy from obscurity to mainstream with amazement, one might ask whether the peak of this development has now been reached or whether the field will continue marching on to new heights. This review looks at the milestones of complement drug discovery and development achieved so far, surveys the currently approved drug entities and indications, and ventures a glimpse into the future advancements yet to come.
Collapse
Affiliation(s)
- Daniel Ricklin
- Molecular Pharmacy Group, Department of Pharmaceutical SciencesUniversity of BaselBaselSwitzerland
| |
Collapse
|
4
|
Li L, Fu R. Advances in research on renal injury in paroxysmal nocturnal hemoglobinuria. J Investig Med 2024; 72:828-833. [PMID: 38715220 DOI: 10.1177/10815589241254046] [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] [Indexed: 08/30/2024]
Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired clonal hematopoietic stem cell disease. Clinical manifestations include intravascular hemolysis, renal dysfunction, fatigue, jaundice, pulmonary hypertension, and so on. Renal injury, as a clinical feature of PNH, is difficult to diagnose and is one of the causes of death in patients with PNH. This article reviews the progress in research on PNH combined with renal injury to improve clinicians' understanding of renal injury in PNH patients, define and judge staging in a timely and accurate manner, enable patients to receive timely and appropriate treatment and reduce mortality.
Collapse
Affiliation(s)
- Liyan Li
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, P.R. China
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, P.R. China
| | - Rong Fu
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, P.R. China
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, P.R. China
| |
Collapse
|
5
|
Schmidt CQ, Höchsmann B, Schrezenmeier H. The complement model disease paroxysmal nocturnal hemoglobinuria. Eur J Immunol 2024; 54:e2350817. [PMID: 39101294 DOI: 10.1002/eji.202350817] [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: 04/02/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/06/2024]
Abstract
We describe initial, current, and future aspects of complement activation and inhibition in the rare hematological disease paroxysmal nocturnal hemoglobinuria (PNH). PNH is a rare but severe hematological disorder characterized by complement-mediated intravascular hemolysis resulting in anemia and severe thrombosis. Insights into the complement-mediated pathophysiology ultimately led to regulatory approval of the first-in-class complement inhibitor, eculizumab, in 2007. This anti-complement C5 therapy resulted in the stabilization of many hematologic parameters and dramatically reduced the often fatal, coagulant-resistant thrombotic events. Despite the remarkable clinical success, a substantial proportion of PNH patients experience suboptimal clinical responses during anti-C5 therapy. We describe the identification and mechanistic dissection of four unexpected processes responsible for such suboptimal clinical responses: (1) pharmacokinetic and (2) pharmacodynamic intravascular breakthrough hemolysis, (3) continuing low-level residual intravascular hemolysis, and (4) extravascular hemolysis. Novel complement therapeutics mainly targeting different complement proteins proximal in the cascade attempt to address these remaining problems. With five approved complement inhibitors in the clinic and many more being evaluated in clinical trials, PNH remains one of the complement diseases with the highest intensity of clinical research. Mechanistically unexpected breakthrough events occur not only with C5 inhibitors but also with proximal pathway inhibitors, which require further mechanistic elaboration.
Collapse
Affiliation(s)
- Christoph Q Schmidt
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Centre, Ulm, Germany
| | - Britta Höchsmann
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden-Württemberg-Hessen and University Hospital of Ulm, Ulm, Germany
| | - Hubert Schrezenmeier
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden-Württemberg-Hessen and University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
6
|
Wang Z, Kulkarni S, Nong J, Zamora M, Ebrahimimojarad A, Hood E, Shuvaeva T, Zaleski M, Gullipalli D, Wolfe E, Espy C, Arguiri E, Wang Y, Marcos-Contreras OA, Song W, Muzykantov VR, Fu J, Radhakrishnan R, Myerson JW, Brenner JS. A percolation-type criticality threshold controls immune protein coating of surfaces. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618530. [PMID: 39464129 PMCID: PMC11507815 DOI: 10.1101/2024.10.15.618530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
When a material enters the body, it is immediately attacked by hundreds of proteins, organized into complex networks of binding interactions and reactions. How do such complex systems interact with a material, "deciding" whether to attack? We focus on the "complement" system of ∼40 blood proteins that bind microbes, nanoparticles, and medical devices, initiating inflammation. We show a sharp threshold for complement activation upon varying a fundamental material parameter, the surface density of potential complement attachment points. This sharp threshold manifests at scales spanning single nanoparticles to macroscale pathologies, shown here for diverse engineered and living materials. Computational models show these behaviors arise from a minimal subnetwork of complement, manifesting percolation-type critical transitions in the complement response. This criticality switch explains the "decision" of a complex signaling network to interact with a material, and elucidates the evolution and engineering of materials interacting with the body.
Collapse
|
7
|
Xiao C, Feng X, Zhao Z, Ding G, Gao Y. Global research trends and focus on the link between neutrophil extracellular traps and tumor: a bibliometric and visualization analysis from 2006 to 2024. Front Immunol 2024; 15:1452104. [PMID: 39381001 PMCID: PMC11459091 DOI: 10.3389/fimmu.2024.1452104] [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: 06/20/2024] [Accepted: 09/13/2024] [Indexed: 10/10/2024] Open
Abstract
Background Neutrophil extracellular traps (NETs) have long been consistently considered an innate immune defense against foreign pathogens, but this oversimplified view has decelerated the progression of perceiving NET biology in chronic diseases. It is now increasingly accepted that NETs are not exclusive to anti-infection responses, but are also central players with a double-edged sword role in cancer progression. NETs have gradually emerged as tumor diagnostic, predictive, and prognostic biomarkers, and strenuous endeavors have been devoted to tapping their potential as new therapeutic targets. Correspondingly, the boom in studies on NETs and tumors in recent years has achieved a series of scientific outputs, which opens up a new perspective for perceiving the sophisticated landscapes of the tumor immune microenvironment. However, there is still much room to translate NET-targeted immunotherapies into clinical practice. Therefore, it is necessary to explore the knowledge structure and latent hotspots of the links between NETs and tumors using bibliometric analysis. Methods NETs and tumor publications from 2006 to 2024 were extracted from the Web of Science Core Collection. Bibliometric analysis and visualization were conducted using Microsoft Excel, VOSviewer, CiteSpace, and R-bibliometrix. Results The analysis included 1,339 publications authored by 7,747 scholars affiliated with 1,926 institutions across 70 countries/regions with relevant articles published in 538 journals. Despite China's maximum number of publications, the United States has continued to dominate the field as a global cooperation center with overwhelming citation counts. Frontiers in Immunology published the most number of publications, whereas Blood was the most cited journal. Wagner, Denisa D. and Kaplan, Mariana J. are concurrently in both the top 10 most prolific authors and cited author lists. Tumor microenvironment and immunotherapy will likely be the focus of future research. Conclusions A comprehensive bibliometric analysis was first conducted to map the current landscape and knowledge structure of the link between NETs and tumors in the hope of providing guidance and fresh perspectives for further research in this field. NETs are promising antitumor targets, and perhaps the eventual destination in the realm is to translate NET-targeted immunotherapies into clinical practice.
Collapse
Affiliation(s)
| | | | | | | | - Yawen Gao
- Department of Oncology, The Second Xiangya Hospital, Central South
University, Changsha, China
| |
Collapse
|
8
|
Stasiłojć M, Stasiłojć G, Kuźniewska A, Rodriguez de Córdoba S, Okrój M. A Cell-Based Assay to Measure the Activity of the Complement Convertases. Kidney Int Rep 2024; 9:2260-2268. [PMID: 39081762 PMCID: PMC11284395 DOI: 10.1016/j.ekir.2024.04.058] [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] [Received: 09/28/2023] [Revised: 04/12/2024] [Accepted: 04/26/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction The complement system serves as a crucial defense mechanism against invading pathogens; however, dysregulation of this system can result in harmful consequences. Central to the complement cascade are the classical pathway (CP) or lectin pathway (LP) and the alternative pathway (AP) convertases. Aberrant regulation of the convertases is often implicated in the development of rare complement-related diseases. However, analyzing convertase activity poses a significant challenge due to their labile nature and intricate interactions with serum proteins. Methods In this study, we propose a novel assay for the functional evaluation of these complexes. Our approach leverages a widely available human lymphoma cell line, which when sensitized with antibodies, triggers activation of the CP with a substantial amplification by the AP. The combined action of 2, C5 blockers eculizumab and crovalimab let the cascade proceed up to the level of convertases but not further. In the next step, C5 inhibitors were washed away and guinea pig serum in ethylenediamine tetraacetic acid (EDTA) buffer supported the development of lytic sites on the platform of preexisting convertases. Results The assay detects recombinant gain-of-function (GoF) components of both convertase types within human serum or plasma. Furthermore, we demonstrate the assay's practical utility in analyzing nephrological patients harboring C3 genetic variants and illustrate its capacity to distinguish between patients and asymptomatic relatives carrying the same pathogenic C3 variant. Conclusion We provided a proof-of-concept of a new assay that detects convertase overactivity in individuals carrying variants of both pathogenic character or those of unknown significance in ubiquitous complement proteins such as C3.
Collapse
Affiliation(s)
- Małgorzata Stasiłojć
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Grzegorz Stasiłojć
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Alicja Kuźniewska
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | | | - Marcin Okrój
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
9
|
Shangguan W, Li X, Wang Y, Huang Z, Dong Y, Feng M, Feng J. Design and Biological Evaluation of the Long-Acting C5-Inhibited Ornithodoros moubata Complement Inhibitor (OmCI) Modified with Fatty Acid. Bioconjug Chem 2024; 35:653-664. [PMID: 38593046 DOI: 10.1021/acs.bioconjchem.4c00126] [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: 04/11/2024]
Abstract
Disorder of complement response is a significant pathogenic factor causing some autoimmune and inflammation diseases. The Ornithodoros moubata Complement Inhibitor (OmCI), a small 17 kDa natural protein, was initially extracted from soft tick salivary glands. The protein was found binding to complement C5 specifically, inhibiting the activation of the complement pathway, which is a successful therapeutic basis of complement-mediated diseases. However, a short half-life due to rapid renal clearance is a common limitation of small proteins for clinical application. In this study, we extended the half-life of OmCI by modifying it with fatty acid, which was a method used to improve the pharmacokinetics of native peptides and proteins. Five OmCI mutants were initially designed, and single-site cysteine mutation was introduced to each of them. After purification, four OmCI mutants were obtained that showed similar in vitro biological activities. Three mutants of them were subsequently coupled with different fatty acids by nucleophilic substitution. In total, 15 modified derivatives were screened and tested for anticomplement activity in vitro. The results showed that coupling with fatty acid would not significantly affect their complement-inhibitory activity (CH50 and AH50). OmCIT90C-CM02 and OmCIT90C-CM05 were validated as the applicable OmCI bioconjugates for further pharmacokinetic assessments, and both showed improved plasma half-life in mice compared with unmodified OmCI (15.86, 17.96 vs 2.57 h). In summary, our data demonstrated that OmCI conjugated with fatty acid could be developed as the potential long-acting C5 complement inhibitor in the clinic.
Collapse
Affiliation(s)
- Wenwen Shangguan
- School of Pharmacy, Fudan University, 201203 Shanghai, China
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Xiaowan Li
- School of Pharmacy, Fudan University, 201203 Shanghai, China
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Yandan Wang
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, 310014 Hangzhou, China
| | - Zongqing Huang
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
- Shanghai Duomirui Biotechnology Co Ltd, 201203 Shanghai, China
| | - Yuanzhen Dong
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
- Shanghai Duomirui Biotechnology Co Ltd, 201203 Shanghai, China
| | - Meiqing Feng
- School of Pharmacy, Fudan University, 201203 Shanghai, China
| | - Jun Feng
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| |
Collapse
|
10
|
Sonnentag SJ, Dopler A, Kleiner K, Garg BK, Mannes M, Späth N, Akilah A, Höchsmann B, Schrezenmeier H, Anliker M, Boyanapalli R, Huber-Lang M, Schmidt CQ. Triple-fusion protein (TriFu): A potent, targeted, enzyme-like inhibitor of all three complement activation pathways. J Biol Chem 2024; 300:105784. [PMID: 38401844 PMCID: PMC11065761 DOI: 10.1016/j.jbc.2024.105784] [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: 07/26/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024] Open
Abstract
The introduction of a therapeutic anti-C5 antibody into clinical practice in 2007 inspired a surge into the development of complement-targeted therapies. This has led to the recent approval of a C3 inhibitory peptide, an antibody directed against C1s and a full pipeline of several complement inhibitors in preclinical and clinical development. However, no inhibitor is available that efficiently inhibits all three complement initiation pathways and targets host cell surface markers as well as complement opsonins. To overcome this, we engineered a novel fusion protein combining selected domains of the three natural complement regulatory proteins decay accelerating factor, factor H and complement receptor 1. Such a triple fusion complement inhibitor (TriFu) was recombinantly expressed and purified alongside multiple variants and its building blocks. We analyzed these proteins for ligand binding affinity and decay acceleration activity by surface plasmon resonance. Additionally, we tested complement inhibition in several in vitro/ex vivo assays using standard classical and alternative pathway restricted hemolysis assays next to hemolysis assays with paroxysmal nocturnal hemoglobinuria erythrocytes. A novel in vitro model of the alternative pathway disease C3 glomerulopathy was established to evaluate the potential of the inhibitors to stop C3 deposition on endothelial cells. Next to the novel engineered triple fusion variants which inactivate complement convertases in an enzyme-like fashion, stoichiometric complement inhibitors targeting C3, C5, factor B, and factor D were tested as comparators. The triple fusion approach yielded a potent complement inhibitor that efficiently inhibits all three complement initiation pathways while targeting to surface markers.
Collapse
Affiliation(s)
- Sophia J Sonnentag
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Centre, Ulm, Germany
| | - Arthur Dopler
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Centre, Ulm, Germany
| | - Katharina Kleiner
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Centre, Ulm, Germany
| | | | - Marco Mannes
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Nadja Späth
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Centre, Ulm, Germany
| | - Amira Akilah
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Centre, Ulm, Germany
| | - Britta Höchsmann
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany; Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden-Württemberg-Hessen and University Hospital of Ulm, Ulm, Germany
| | - Hubert Schrezenmeier
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany; Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden-Württemberg-Hessen and University Hospital of Ulm, Ulm, Germany
| | - Markus Anliker
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany; Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden-Württemberg-Hessen and University Hospital of Ulm, Ulm, Germany
| | | | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Christoph Q Schmidt
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Centre, Ulm, Germany; Institute of Pharmacy, Biochemical Pharmacy Group, Martin Luther University Halle-Wittenberg, Halle, Germany.
| |
Collapse
|
11
|
Hu Y, Wang H, Liu Y. NETosis: Sculpting tumor metastasis and immunotherapy. Immunol Rev 2024; 321:263-279. [PMID: 37712361 DOI: 10.1111/imr.13277] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/16/2023]
Abstract
The process of neutrophil extracellular traps (NETs) formation, called NETosis, is a peculiar death modality of neutrophils, which was first observed as an immune response against bacterial infection. However, recent work has revealed the unique biology of NETosis in facilitating tumor metastatic process. Neutrophil extracellular traps released by the tumor microenvironment (TME) shield tumor cells from cytotoxic immunity, leading to impaired tumor clearance. Besides, tumor cells tapped by NETs enable to travel through vessels and subsequently seed distant organs. Targeted ablation of NETosis has been proven to be beneficial in potentiating the efficacy of cancer immunotherapy in the metastatic settings. This review outlines the impact of NETosis at almost all stages of tumor metastasis. Furthermore, understanding the multifaceted interplay between NETosis and the TME components is crucial for supporting the rational development of highly effective combination immunotherapeutic strategies with anti-NETosis for patients with metastatic disease.
Collapse
Affiliation(s)
- Yanyan Hu
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Houhong Wang
- Department of General Surgery, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, China
| | - Yang Liu
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
12
|
Fishman J, Anderson S, Talbird SE, Dingli D. Analysis of Costs per Responder in US Adults with Paroxysmal Nocturnal Hemoglobinuria with a Suboptimal Response to Prior Eculizumab Treatment. Hematol Rep 2023; 15:578-591. [PMID: 37873795 PMCID: PMC10594490 DOI: 10.3390/hematolrep15040060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/06/2023] [Indexed: 10/25/2023] Open
Abstract
European Society for Blood and Marrow Transplantation (EBMT) hematologic response categories comprehensively assess complement inhibitor responses in patients with paroxysmal nocturnal hemoglobinuria (PNH). Using data from the 16-week randomized controlled period of the phase 3 PEGASUS trial (N = 80), we estimated the treatment cost per responder by the EBMT response category for pegcetacoplan and eculizumab in adults with PNH and a suboptimal response to eculizumab. Average drug costs per responder, number needed to treat, and incremental drug costs per responder were estimated using dosages administered during the trial (base case). A US payer perspective (2020 US dollars) was used. Scenario analyses were conducted for various costs, dosages, treatment durations, patient populations, and settings. In total, 30 of 41 (73%) who switched to pegcetacoplan and 2 of 39 (5%) patients who continued eculizumab had a good, major, or complete response (good-to-complete responders) at Week 16. Average weekly drug costs per good-to-complete responder were USD 15,923 with pegcetacoplan and USD 216,100 with eculizumab; average weekly drug costs per patient were USD 11,651 and USD 11,082, respectively. Average drug costs per good-to-complete responder with pegcetacoplan were similar across complement inhibitor-naïve populations and were consistently lower than with eculizumab. Switching from eculizumab to pegcetacoplan allowed more patients with a suboptimal response to attain a good-to-complete response at lower costs. These results apply to patients with a suboptimal response to prior eculizumab treatment only.
Collapse
Affiliation(s)
- Jesse Fishman
- Apellis Pharmaceuticals, Inc., Waltham, MA 02451, USA;
| | - Seri Anderson
- RTI Health Solutions, Research Triangle Park, NC 27709, USA;
| | | | - David Dingli
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| |
Collapse
|
13
|
Bortolotti M, Barcellini W, Fattizzo B. Molecular pharmacology in complement-mediated hemolytic disorders. Eur J Haematol 2023. [PMID: 37308291 DOI: 10.1111/ejh.14026] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/14/2023]
Abstract
In the last decade, a deeper understanding of the pathogenesis of complement mediated hemolytic disorders, such as paroxysmal nocturnal hemoglobinuria (PNH), cold agglutinin disease (CAD), warm type autoimmune hemolytic anemia (AIHA) with complement activation (wAIHA), and atypical hemolytic uremic syndrome (aHUS), paved the way to the therapeutic shift from purely supportive approaches to complement-targeted therapies. This resulted in a significant improvement in disease management, survival, and quality of life. In this review, we will provide a snapshot of novel therapies for complement-mediated hemolytic anemias with a focus on those ready to use in clinical practice. C5 inhibitors eculizumab and the long-acting ravulizumab, are the established gold standard for untreated PNH patients, whilst the C3 inhibitor pegcetacoplan should be considered for suboptimal responders to anti-C5 drugs. Several additional compounds targeting the complement cascade at different levels (other C5 inhibitors, factor B and D inhibitors) are under active investigation with promising results. In CAD, immunosuppression with rituximab remains the first-line. However, recently FDA and EMA approved the anti-C1s monoclonal antibody, sutimlimab, that showed dramatic responses and whose regulatory approval is soon awaited in many countries. Other drugs under investigation in AIHA include the C3 inhibitor pegcetacoplan, and the anti-C1q ANX005 for warm AIHA with complement activation. Finally, aHUS is an indication for complement inhibitors. Eculizumab and ravulizumab have been approved, whilst other C5 inhibitors, and novel lectin pathway inhibitors are under active investigation in this disease.
Collapse
Affiliation(s)
- Marta Bortolotti
- Hematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Oncohematology, University of Milan, Milan, Italy
| | - Wilma Barcellini
- Hematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Bruno Fattizzo
- Hematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Oncohematology, University of Milan, Milan, Italy
| |
Collapse
|
14
|
Dong J, Zheng F, Liu F, He J, Li S, Pu W, Xu H, Luo Z, Liu S, Yin L, Tang D, Dai Y. Global-feature of autoimmune glomerulonephritis using proteomic analysis of laser capture microdissected glomeruli. Front Immunol 2023; 14:1131164. [PMID: 37033921 PMCID: PMC10077062 DOI: 10.3389/fimmu.2023.1131164] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 02/13/2023] [Indexed: 04/11/2023] Open
Abstract
Background IgA nephropathy (IgAN), (LN), membranous nephropathy (MN), and minimal change nephropathy (MCN) are all belonged to autoimmune glomerulonephritis. This study aimed to identify the specific proteomic characteristics of the four GNs diseases in order to provide frameworks for developing the appropriate drug for patients diagnosed with GNs disease. Methods Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was utilized to investigate proteomic features of glomerular tissues obtained by laser capture microdissection (LCM). 8 normal control cases, 11 IgAN cases, 19 LN cases, 5 MN cases, and 3 MCN cases in this study were selected for bioinformatics analyses. Results The shared overlapping proteins among the top 100 DEPs of each GNs type were mostly downregulated, in which only FLII was significantly downregulated in the four GNs diseases. A2M was significantly upregulated in MN, IgAN, and LN subgroups. The pathway of complement and coagulation cascades was notably activated with NES value ranging 2.77 to 3.39 among MCN, MN, IgAN, and LN diseases, but the pattern of protein expression level were significantly different. In LN patients, the increased activity of complement and coagulation cascades was contributed by the high expression of multiple complements (C1QB, C3, C4A, C4B, C6, C8B, C8G, C9). Meanwhile, both C1QC and C4B were remarkably upregulated in MN patients. On the contrary, complement-regulating proteins (CD59) was substantially decreased in MCN and IgAN subgroup. Conclusions The integrative proteomics analysis of the four GNs diseases provide insights into unique characteristics of GNs diseases and further serve as frameworks for precision medicine diagnosis and provide novel targets for drug development.
Collapse
Affiliation(s)
- Jingjing Dong
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Fengping Zheng
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Fanna Liu
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Jingquan He
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Shanshan Li
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Wenjun Pu
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Huixuan Xu
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Zhifeng Luo
- Guangxi Key Laboratory of Metabolic Disease Research, The 924th Hospital of the Chinese People’s Liberation Army Joint Logistic Support Force, Guilin, Guangxi, China
| | - Shizhen Liu
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Lianghong Yin
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- *Correspondence: Lianghong Yin, ; Donge Tang, ; Yong Dai,
| | - Donge Tang
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Lianghong Yin, ; Donge Tang, ; Yong Dai,
| | - Yong Dai
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
- Guangxi Key Laboratory of Metabolic Disease Research, The 924th Hospital of the Chinese People’s Liberation Army Joint Logistic Support Force, Guilin, Guangxi, China
- *Correspondence: Lianghong Yin, ; Donge Tang, ; Yong Dai,
| |
Collapse
|