Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 288
Filter
1.
Nephrology (Carlton) ; 29 Suppl 2: 44-46, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39327741

ABSTRACT

The role of complement in the pathogenesis of IgA nephropathy has been heavily explored over the past 50 years. This has led to the general acceptance that complement plays an important role in the clinical presentation and risk for progression of disease in patients with IgA nephropathy. Herein, we review the evidence for complement activation in IgA nephropathy, focusing on evidence that the lectin and alternate pathways are the main actors. We are entering an era of intense investigation of various inhibitors of complement, which should ultimately be the best indicator of contributions of the lectin, alternate and common complement pathways to disease burden. More importantly, we will see if these efforts result in the discovery of clinically relevant options in managing this important disease.


Subject(s)
Complement Activation , Complement Inactivating Agents , Glomerulonephritis, IGA , Glomerulonephritis, IGA/immunology , Glomerulonephritis, IGA/drug therapy , Humans , Complement Inactivating Agents/therapeutic use , Complement Inactivating Agents/pharmacology , Complement Activation/drug effects , Complement System Proteins/immunology , Complement System Proteins/metabolism , Animals , Complement Pathway, Alternative/drug effects , Complement Pathway, Alternative/immunology
2.
Carbohydr Polym ; 343: 122448, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39174088

ABSTRACT

Fucoidans, a group of high molecular weight polysaccharides derived mainly from brown algae, are characterized by their high fucose content, degree of sulfation (DS), and intra- and interspecific structural variation. Fucoidans are increasingly recognized due to various reported bioactivities, potentially beneficial for human health. To unlock their potential use within biomedical applications, a better understanding of their structure-functional relationship is needed. To achieve this, systematic bioactivity studies based on well-defined, pure fucoidans, and the establishment of standardized, satisfactory purification protocols are required. We performed a comprehensive compositional and structural characterization of crude and ultra-purified fucoidans from three kelps: Saccharina latissima (SL), Alaria esculenta (AE) and Laminaria hyperborea (LH). Further, the complement-inhibiting activity of the purified fucoidans was assessed in a human whole blood model. The purification process led to fucoidans with higher DS and fucose and lower concentrations of other monosaccharides. Fucoidans from SL and LH resembles homofucans, while AE is a heterofucan rich in galactose with comparably lower DS. Fucoidans from SL and LH showed complement-inhibiting activity in blood and blood plasma, while no inhibition was observed for AE under the same conditions. The results emphasize the importance of high DS and possibly fucose content for fucoidans' bioactive properties.


Subject(s)
Edible Seaweeds , Kelp , Laminaria , Phaeophyceae , Polysaccharides , Humans , Complement Inactivating Agents/chemistry , Complement Inactivating Agents/isolation & purification , Complement Inactivating Agents/pharmacology , Edible Seaweeds/chemistry , Fucose/chemistry , Immunologic Factors/pharmacology , Immunologic Factors/chemistry , Immunologic Factors/isolation & purification , Kelp/chemistry , Laminaria/chemistry , Phaeophyceae/chemistry , Polysaccharides/chemistry , Polysaccharides/pharmacology , Polysaccharides/isolation & purification , Water/chemistry
3.
J Peripher Nerv Syst ; 29(3): 339-349, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38987228

ABSTRACT

BACKGROUND AND AIMS: Guillain-Barré syndrome (GBS) is an acute, self-limited, immune-mediated peripheral neuropathy. Current treatments for GBS include intravenous immunoglobulin (IVIg) and plasma exchange, which may not sufficiently benefit severely affected patients. This study evaluated the efficacy and safety of eculizumab add-on therapy to IVIg (standard-of-care treatment) in patients with severe GBS. METHODS: This phase 3, multicenter, double-blind, randomized, placebo-controlled clinical trial (NCT04752566), enrolled Japanese adults (age ≥ 18 years) with severe GBS (Hughes functional grade [FG] score FG3 or FG4/FG5 within 2 weeks of onset of GBS). Participants were randomized 2:1 to receive intravenous infusion of eculizumab or placebo (once weekly for 4 weeks) with IVIg treatment with 20 weeks of follow-up. Primary efficacy endpoint was the time to first reach FG score ≤1 (able to run). Key secondary endpoints were proportion of participants achieving FG ≤1 at weeks 8 and 24 and FG improvement ≥3 at week 24. Pharmacodynamic analysis of serum free C5 concentration over time was performed. Safety was evaluated. RESULTS: The analysis included 57 participants (eculizumab, n = 37; placebo, n = 20). Primary endpoint was not achieved (hazard ratio, 0.9; 95% CI, 0.45-1.97; p = .89). Key secondary endpoints did not reach statistical significance. Serum C5 concentration was reduced by 99.99% at 1 h postdose and sustained to week 5 but returned to baseline at the end of follow-up period. No new safety signals for eculizumab were identified. INTERPRETATION: Although well tolerated, eculizumab treatment did not show significant effects on motor function recovery compared to placebo in patients with GBS.


Subject(s)
Antibodies, Monoclonal, Humanized , Guillain-Barre Syndrome , Humans , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacology , Male , Guillain-Barre Syndrome/drug therapy , Guillain-Barre Syndrome/physiopathology , Double-Blind Method , Female , Middle Aged , Adult , Aged , Immunoglobulins, Intravenous/administration & dosage , Immunoglobulins, Intravenous/adverse effects , Complement Inactivating Agents/administration & dosage , Complement Inactivating Agents/pharmacology , Complement Inactivating Agents/adverse effects , Treatment Outcome
4.
J Neurol ; 271(9): 6209-6219, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39080054

ABSTRACT

INTRODUCTION: Eculizumab, a complement active antibody, and efgartigimod, an Fc fragment that blocks neonatal Fc receptor, are both approved to treat generalized myasthenia gravis (gMG) patients. The objective of this study is to describe the clinical response to both treatments in a real-life setting. METHODS: We collected baseline and follow-up clinical data using the Myasthenia Gravis-Activities of Daily Living (MG-ADL), and Quantitative Myasthenia Gravis (QMG). We included 63 patients, 32 treated with eculizumab and 31 with efgartigimod. Of the efgartigimod-treated patients, 22 were anti-acetylcholine receptor antibody-positive (AChR-Ab +) and 9 were AChR-Ab- (3 MuSK-Ab + and 6 seronegative). RESULTS: Both treatments showed similar efficacy relative to the MG-ADL scale reduction (p = 0.237). Efgartigimod had a similar effect on both AChR-Ab + and AChR-Ab- (p = 0.280). Eculizumab was superior to efgartigimod relative to the QMG score reduction for the entire dataset (p = 0.003) and was more likely to achieve a clinical response at the QMG compared to efgartigimod (OR 1.373; p = 0.016). Steroid-sparing effect was higher for eculizumab than efgartigimod ( - 16.7 vs - 5.2 mg of the baseline daily dose at follow-up; p = 0.001). Mean speed of prednisone reduction was - 13.1 mg of the daily dose for each month of follow-up for eculizumab-treated patients and - 3.2 for efgartigimod (p = 0.001). We found three serious events, all not related to treatment in the investigator's opinion. One eculizumab-treated patient experienced a severe pneumonia and died despite treatment. CONCLUSIONS: Our study provides evidence that eculizumab and efgartigimod can be used in clinical practice to reduce disability in gMG patients. Eculizumab-treated patients had a higher QMG response and steroid sparing effect. Efgartigimod may offer a more flexible schedule due to its cyclical use, no need for vaccination, and efficacy in AChR-Ab- patients.


Subject(s)
Antibodies, Monoclonal, Humanized , Myasthenia Gravis , Humans , Myasthenia Gravis/drug therapy , Female , Male , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacology , Middle Aged , Adult , Aged , Treatment Outcome , Receptors, Cholinergic/immunology , Complement Inactivating Agents/pharmacology , Complement Inactivating Agents/administration & dosage , Activities of Daily Living , Follow-Up Studies
6.
J Biol Chem ; 300(7): 107467, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38876307

ABSTRACT

The complement system plays a critical role in the innate immune response, acting as a first line of defense against invading pathogens. However, dysregulation of the complement system is implicated in the pathogenesis of numerous diseases, ranging from Alzheimer's to age-related macular degeneration and rare blood disorders. As such, complement inhibitors have enormous potential to alleviate disease burden. While a few complement inhibitors are in clinical use, there is still a significant unmet medical need for the discovery and development of novel inhibitors to treat patients suffering from disorders of the complement system. A key hurdle in the development of complement inhibitors has been the determination of their mechanism of action. Progression along the complement cascade involves the formation of numerous multimeric protein complexes, creating the potential for inhibitors to act at multiple nodes in the pathway. This is especially true for molecules that target the central component C3 and its fragment C3b, which serve a dual role as a substrate for the C3 convertases and as a scaffolding protein in both the C3 and C5 convertases. Here, we report a step-by-step in vitro reconstitution of the complement alternative pathway using bio-layer interferometry. By physically uncoupling each step in the pathway, we were able to determine the kinetic signature of inhibitors that act at single steps in the pathway and delineate the full mechanism of action of known and novel C3 inhibitors. The method could have utility in drug discovery and further elucidating the biochemistry of the complement system.


Subject(s)
Complement Pathway, Alternative , Humans , Complement Pathway, Alternative/drug effects , Complement C3/metabolism , Complement C3/antagonists & inhibitors , Complement Inactivating Agents/pharmacology , Complement C3b/metabolism
7.
Blood Adv ; 8(15): 4181-4193, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38865712

ABSTRACT

ABSTRACT: Excessively activated or dysregulated complement activation may contribute to the pathogenesis of a wide range of human diseases, thus leading to a surge in complement inhibitors. Herein, we developed a human-derived and antibody-like C3b-targeted fusion protein (CRIg-FH-Fc) x2, termed CG001, that could potently block all 3 complement pathways. Complement receptor of the immunoglobulin superfamily (CRIg) and factor H (FH) bind to distinct sites in C3b and synergistically inhibit complement activation. CRIg occupancy in C3b prevents the recruitment of C3 and C5 substrates, whereas FH occupancy in C3b accelerates the decay of C3/C5 convertases and promotes the factor I-mediated degradation and inactivation of C3b. CG001 also showed therapeutic effects in alternative pathways-induced hemolytic mouse and classical pathways-induced mesangial proliferative glomerulonephritis rat models. In the pharmacological/toxicological evaluation in rats and cynomolgus monkeys, CG001 displayed an antibody-like pharmacokinetic profile, a convincing complement inhibitory effect, and no observable toxic effects. Therefore, CG001 holds substantial potential for human clinical studies.


Subject(s)
Complement C3b , Animals , Humans , Rats , Mice , Complement C3b/metabolism , Complement Inactivating Agents/pharmacology , Complement Inactivating Agents/therapeutic use , Macaca fascicularis , Complement Activation/drug effects , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Complement Factor H/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical
8.
Medicina (Kaunas) ; 60(6)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38929562

ABSTRACT

The complement cascade is a vital system in the human body's defense against pathogens. During the natural aging process, it has been observed that this system is imperative for ensuring the integrity and homeostasis of the retina. While this system is critical for proper host defense and retinal integrity, it has also been found that dysregulation of this system may lead to certain retinal pathologies, including geographic atrophy and diabetic retinopathy. Targeting components of the complement system for retinal diseases has been an area of interest, and in vivo, ex vivo, and clinical trials have been conducted in this area. Following clinical trials, medications targeting the complement system for retinal disease have also become available. In this manuscript, we discuss the pathophysiology of complement dysfunction in the retina and specific pathologies. We then describe the results of cellular, animal, and clinical studies targeting the complement system for retinal diseases. We then provide an overview of complement inhibitors that have been approved by the Food and Drug Administration (FDA) for geographic atrophy. The complement system in retinal diseases continues to serve as an emerging therapeutic target, and further research in this field will provide additional insights into the mechanisms and considerations for treatment of retinal pathologies.


Subject(s)
Complement System Proteins , Retinal Diseases , Humans , Retinal Diseases/drug therapy , Retinal Diseases/physiopathology , Retinal Diseases/immunology , Complement System Proteins/physiology , Animals , Complement Inactivating Agents/therapeutic use , Complement Inactivating Agents/pharmacology , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/physiopathology , Retina/drug effects , Retina/immunology
9.
Pediatr Neurol ; 156: 198-207, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38810600

ABSTRACT

BACKGROUND: This study evaluated the efficacy and safety of eculizumab, a terminal complement C5 inhibitor, in juvenile generalized myasthenia gravis (gMG). METHODS: Adolescents aged 12 to 17 years with refractory anti-acetylcholine receptor (AChR) antibody-positive gMG received eculizumab (weekly induction [one to two doses of 600 mg or four doses of 900 mg] followed by maintenance doses [300 to 1200 mg] every two weeks for up to 26 weeks) in a phase 3, open-label multicenter study (NCT03759366). Change from baseline to week 26 in Quantitative Myasthenia Gravis (QMG) total score (primary end point) and secondary end points including Myasthenia Gravis-Activities of Daily Living (MG-ADL) total score, Myasthenia Gravis Composite score, Myasthenia Gravis Foundation of America postintervention status, EuroQol 5-Dimensions (Youth) and Neurological Quality-of-Life Pediatric Fatigue questionnaire scores, as well as pharmacokinetics, pharmacodynamics, and safety, were recorded. RESULTS: Eleven adolescents (mean ± S.D. age 14.8 ± 1.8 years) were enrolled; 10 completed the primary evaluation period. Least-squares mean changes from baseline at week 26 were -5.8 (standard error [SE] 1.2; P = 0.0004) for QMG total score and -2.3 (SE 0.6; P = 0.0017) for MG-ADL total score. Overall, the primary and all secondary efficacy end point analyses met statistical significance from the first assessment and were sustained throughout. Complete terminal complement inhibition was sustained through 26 weeks in all patients. Treatment-emergent adverse events were all mild/moderate and predominantly unrelated to eculizumab. CONCLUSIONS: Eculizumab was effective in reducing disease burden and was well tolerated in adolescents with refractory AChR antibody-positive gMG.


Subject(s)
Antibodies, Monoclonal, Humanized , Complement Inactivating Agents , Myasthenia Gravis , Humans , Adolescent , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacology , Myasthenia Gravis/drug therapy , Male , Female , Child , Complement Inactivating Agents/administration & dosage , Complement Inactivating Agents/pharmacology , Treatment Outcome , Quality of Life , Outcome Assessment, Health Care
10.
Drugs ; 84(6): 707-716, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38740735

ABSTRACT

Crovalimab (®; PiaSky) is a humanized, anti-complement component C5 (anti-C5) recycling monoclonal antibody developed by Chugai Pharmaceutical, in collaboration with Roche, which is being investigated for the treatment of complement-mediated diseases, including paroxysmal nocturnal haemoglobinuria (PNH), atypical haemolytic uremic syndrome, lupus nephritis and sickle cell disease. Crovalimab targets C5, inhibiting its cleavage to C5a and C5b, thus blocking the terminal complement pathway and preventing intravascular haemolysis in PNH. Crovalimab is designed to bind to the antigen repeatedly, resulting in sustained complement inhibition at a lower dosage, and allowing for once-monthly subcutaneous administration. In February 2024, subcutaneous crovalimab received its first approval in China for the treatment of adolescents and adults (aged ≥ 12 years) with PNH who have not been previously treated with complement inhibitors. Crovalimab has since been approved in Japan in March for use in the treatment of PNH, including in treatment-naïve and previously treated patients. Crovalimab is also under regulatory review for the treatment of naïve and previously treated patients with PNH in multiple countries, including the USA and the European Union. This article summarizes the milestones in the development of crovalimab leading to this first approval in China for the treatment of PNH.


Subject(s)
Antibodies, Monoclonal, Humanized , Drug Approval , Hemoglobinuria, Paroxysmal , Humans , Hemoglobinuria, Paroxysmal/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/adverse effects , Complement C5/antagonists & inhibitors , China , Adult , Adolescent , Complement Inactivating Agents/therapeutic use , Complement Inactivating Agents/pharmacology , Atypical Hemolytic Uremic Syndrome/drug therapy
11.
Ophthalmologie ; 121(6): 476-481, 2024 Jun.
Article in German | MEDLINE | ID: mdl-38691156

ABSTRACT

The approval of complement inhibitory therapeutic agents for the treatment of geographic atrophy (GA) has highlighted the need for reliable and reproducible measurement of disease progression and therapeutic efficacy. Due to its availability and imaging characteristics optical coherence tomography (OCT) is the method of choice. Using OCT analysis based on artificial intelligence (AI), the therapeutic efficacy of pegcetacoplan was demonstrated at the levels of both the retinal pigment epithelium (RPE) and photoreceptors (PR). Cloud-based solutions that enable monitoring of GA are already available.


Subject(s)
Biomarkers , Complement Inactivating Agents , Geographic Atrophy , Tomography, Optical Coherence , Humans , Geographic Atrophy/drug therapy , Geographic Atrophy/metabolism , Complement Inactivating Agents/therapeutic use , Complement Inactivating Agents/pharmacology , Treatment Outcome , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/diagnostic imaging , Retinal Pigment Epithelium/metabolism
12.
Bioconjug Chem ; 35(5): 653-664, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38593046

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.


Subject(s)
Complement C5 , Fatty Acids , Ornithodoros , Animals , Fatty Acids/chemistry , Mice , Complement C5/antagonists & inhibitors , Drug Design , Half-Life , Complement Inactivator Proteins/pharmacology , Complement Inactivator Proteins/chemistry , Complement Inactivating Agents/pharmacology , Complement Inactivating Agents/pharmacokinetics , Complement Inactivating Agents/chemistry , Humans
13.
Ann Clin Transl Neurol ; 11(5): 1338-1346, 2024 May.
Article in English | MEDLINE | ID: mdl-38572524

ABSTRACT

OBJECTIVE: Eculizumab and ravulizumab are complement protein C5 inhibitors, showing efficacy and tolerability for patients with anti-acetylcholine receptor-positive (AChR+) generalized myasthenia gravis (gMG) in phase 3 clinical trials and subsequent analyses. The purpose of the present study was to evaluate the clinical significance of eculizumab and switching to ravulizumab for refractory AChR+ gMG patients in the real-world experience. METHODS: Among the database of Japan MG registry survey 2021, we studied AChR+ gMG patients who received eculizumab. We also evaluated these patients who switched from eculizumab to ravulizumab. Responder was defined as an improvement of at least 3 points in MG-ADL. We performed a questionnaire of preference between eculizumab and ravulizumab. RESULTS: Among 1,106 patients with AChR+ gMG, 36 patients (3%) received eculizumab (female 78%, mean age 56.0 years). Eculizumab was preferentially used in severe and refractory MG patients. The duration of eculizumab treatment was 35 months on average. MG-ADL improved from 9.4 ± 4.9 to 5.9 ± 5.1, and 25 (70%) of the 36 gMG patients were responders. Postintervention status was markedly improved after the eculizumab treatment. Of 13 patients who did not continue eculizumab, 6 showed insufficiencies. Early onset MG was most effective. However, 15 patients switching from eculizumab to ravulizumab kept favorable response and tolerability. Questionnaire surveys showed preference for ravulizumab over eculizumab. INTERPRETATION: Eculizumab and switching to ravulizumab showed to be effective for refractory AChR+ gMG patients in clinical settings.


Subject(s)
Antibodies, Monoclonal, Humanized , Complement Inactivating Agents , Myasthenia Gravis , Humans , Myasthenia Gravis/drug therapy , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Female , Male , Middle Aged , Aged , Adult , Complement Inactivating Agents/administration & dosage , Complement Inactivating Agents/pharmacology , Drug Substitution , Registries , Japan
14.
Drugs ; 84(5): 599-606, 2024 May.
Article in English | MEDLINE | ID: mdl-38517653

ABSTRACT

Iptacopan (FABHALTA®) is an oral complement Factor B inhibitor developed by Novartis Pharmaceuticals for the treatment of complement-mediated diseases. Acting upstream of complement 5 in the alternative pathway, iptacopan inhibits both terminal complement-mediated intravascular haemolysis and complement 3-mediated extravascular haemolysis. On 5 December 2023, iptacopan received approval in the USA for the treatment of adults with paroxysmal nocturnal haemoglobinuria (PNH). This article summarizes the milestones in the development of iptacopan leading to this first approval for PNH.


Subject(s)
Drug Approval , Hemoglobinuria, Paroxysmal , Humans , Complement Inactivating Agents/therapeutic use , Complement Inactivating Agents/pharmacology , Hemoglobinuria, Paroxysmal/drug therapy , Hemolysis/drug effects , United States , United States Food and Drug Administration
15.
Drugs ; 84(5): 613-618, 2024 May.
Article in English | MEDLINE | ID: mdl-38528310

ABSTRACT

Danicopan (Voydeya®) is an oral complement factor D inhibitor that is being developed by Alexion AstraZeneca Rare Disease as add-on treatment to ravulizumab or eculizumab for patients with clinically significant extravascular haemolysis. Danicopan recently received approval in Japan for the treatment of adults with paroxysmal nocturnal haemoglobinuria (PNH) when used in addition to a complement component 5 (C5) inhibitor. Subsequently, the European Medicines Agency adopted a positive opinion recommending the granting of marketing authorisation for danicopan for the treatment of patients with PNH who continue to have residual haemolytic anaemia despite treatment with a complement C5 inhibitor. This article summarizes the milestones in the development of danicopan leading to this first approval for PNH.


Subject(s)
Antibodies, Monoclonal, Humanized , Drug Approval , Hemoglobinuria, Paroxysmal , Humans , Hemoglobinuria, Paroxysmal/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/adverse effects , Complement Inactivating Agents/therapeutic use , Complement Inactivating Agents/pharmacology
16.
Eur J Intern Med ; 124: 22-31, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38461065

ABSTRACT

The Complement System is an evolutionarily conserved component of immunity that plays a key role in host defense against infections and tissue homeostasis. However, the dysfunction of the Complement System can result in tissue damage and inflammation, thereby contributing to the development and progression of various renal diseases, ranging from atypical Hemolytic Uremic Syndrome to glomerulonephritis. Therapeutic interventions targeting the complement system have demonstrated promising results in both preclinical and clinical studies. Currently, several complement inhibitors are being developed for the treatment of complement-mediated renal diseases. This review aims to summarize the most recent insights into complement activation and therapeutic inhibition in renal diseases. Furthermore, it offers potential directions for the future rational use of complement inhibitor drugs in the context of renal diseases.


Subject(s)
Complement Activation , Complement Inactivating Agents , Complement System Proteins , Disease Progression , Kidney Diseases , Humans , Complement System Proteins/immunology , Kidney Diseases/immunology , Kidney Diseases/etiology , Complement Inactivating Agents/therapeutic use , Complement Inactivating Agents/pharmacology , Animals , Atypical Hemolytic Uremic Syndrome/immunology , Glomerulonephritis/immunology
17.
Expert Opin Pharmacother ; 25(4): 395-408, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38523508

ABSTRACT

INTRODUCTION: Myasthenia gravis (MG) is an autoimmune condition targeting the neuromuscular junction, which manifests with neuromuscular symptoms of varying severity and significant morbidity. The mainstay of treatment in MG is mitigation of the immune cascade with steroids and non-steroidal immunosuppressive therapies. The therapeutic strategies in MG are transitioning from broad and indiscriminate immunosuppression to novel agents targeting key steps in MG pathogenesis, including T cell activation, B cell proliferation, complement activation, maintenance of pathogenic antibody production, and proinflammatory cytokine production. AREAS COVERED: In this review, an overview of the pathogenesis of MG and traditional MG therapies is presented, followed by a discussion of the novel MG drugs that have been evaluated in phase 3 clinical trials with an emphasis on those which have received regulatory approval. EXPERT OPINION: Novel MG therapeutics belonging to the classes of complement inhibitors, neonatal Fc receptor (FcRn) inhibitors and B cell depletors, as well as the other emerging MG drugs in the pipeline constitute promising treatment strategies with potentially better efficacy and safety compared to the conventional MG treatments. However, further long-term research is needed in order to optimize the implementation of these new treatment options for the appropriate patient populations.


Subject(s)
Immunosuppressive Agents , Myasthenia Gravis , Humans , Myasthenia Gravis/drug therapy , Myasthenia Gravis/immunology , Myasthenia Gravis/therapy , Immunosuppressive Agents/therapeutic use , Animals , B-Lymphocytes/immunology , B-Lymphocytes/drug effects , Complement Inactivating Agents/therapeutic use , Complement Inactivating Agents/pharmacology
18.
J Drug Target ; 32(5): 485-498, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38491993

ABSTRACT

The complement is an important arm of the innate immune system, once activated, the complement system rapidly generates large quantities of protein fragments that are potent mediators of inflammation. Recent studies have shown that over-activated complement is the main proinflammatory system of autoimmune diseases (ADs). In addition, activated complements interact with autoantibodies, immune cells exacerbate inflammation, further worsening ADs. With the increasing threat of ADs to human health, complement-based immunotherapy has attracted wide attention. Nevertheless, efficient and targeted delivery of complement inhibitors remains a significant challenge owing to their inherent poor targeting, degradability, and low bioavailability. Nanosystems offer innovative solutions to surmount these obstacles and amplify the potency of complement inhibitors. This prime aim to present the current knowledge of complement in ADs, analyse the function of complement in the pathogenesis and treatment of ADs, we underscore the current situation of nanosystems assisting complement inhibitors in the treatment of ADs. Considering technological, physiological, and clinical validation challenges, we critically appraise the challenges for successfully translating the findings of preclinical studies of these nanosystem assisted-complement inhibitors into the clinic, and future perspectives were also summarised. (The graphical abstract is by BioRender.).


Subject(s)
Autoimmune Diseases , Complement Inactivating Agents , Complement System Proteins , Humans , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Complement Inactivating Agents/administration & dosage , Complement Inactivating Agents/pharmacology , Animals , Complement System Proteins/immunology , Nanoparticles , Complement Activation/drug effects , Drug Delivery Systems , Immunotherapy/methods
20.
J Biol Chem ; 300(4): 105784, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38401844

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.


Subject(s)
Complement Factor H , Receptors, Complement 3b , Recombinant Fusion Proteins , Humans , Complement Factor H/metabolism , Complement Factor H/genetics , Complement Factor H/chemistry , Complement Factor H/immunology , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Complement Activation/drug effects , CD55 Antigens/genetics , CD55 Antigens/metabolism , Hemolysis/drug effects , Complement Pathway, Alternative/drug effects , Complement Inactivating Agents/pharmacology , Erythrocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL