Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
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
2.
Blood Adv ; 8(11): 2813-2824, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38551806

ABSTRACT

ABSTRACT: Paroxysmal nocturnal hemoglobinuria (PNH) is a rare hematologic disease of uncontrolled terminal complement activation leading to intravascular hemolysis, thrombotic events and increased morbidity and mortality. This phase 3, open-label, single-arm, multicenter study evaluated ravulizumab treatment in eculizumab-naive or -experienced pediatric patients (aged <18 years) with PNH over a 26-week primary evaluation period (PEP) and 4-year extension period (EP). Patients included in the study received weight-based intravenous ravulizumab dosing. Primary end points were pharmacokinetic and pharmacodynamic parameters to confirm complement component 5 (C5) inhibition by ravulizumab; secondary end points assessed the efficacy (including percentage change in lactate dehydrogenase levels over time) and safety of ravulizumab. Thirteen patients, 5 (38.5%) eculizumab-naive and 8 (61.5%) eculizumab-experienced, were enrolled. Ravulizumab Ctrough levels were above the pharmacokinetic threshold of 175 µg/mL in the PEP and EP except in 1 patient. At the end of the study, pre- and post-infusion mean ± standard deviation serum ravulizumab concentrations were 610.50 ± 201.53 µg/mL and 518.29 ± 109.67 µg/mL for eculizumab-naive and eculizumab-experienced patients, respectively. After the first ravulizumab infusion, serum-free C5 concentrations were <0.5 µg/mL in both cohorts until the end of the study (0.061 ± 0.021 µg/mL and 0.061 ± 0.018 µg/mL for eculizumab-naive and eculizumab-experienced patients, respectively). Compared with baseline, ravulizumab improved and maintained efficacy outcomes in both groups. Ravulizumab had an acceptable safety profile with no new safety signals identified, and provided immediate, complete, and sustained terminal complement inhibition, translating to clinical benefit for pediatric patients with PNH. This trial was registered at www.ClinicalTrials.gov as #NCT03406507.


Subject(s)
Antibodies, Monoclonal, Humanized , Hemoglobinuria, Paroxysmal , Humans , Hemoglobinuria, Paroxysmal/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Child , Female , Male , Adolescent , Treatment Outcome , Child, Preschool , Complement Inactivating Agents/pharmacokinetics , Complement Inactivating Agents/therapeutic use , Complement Inactivating Agents/adverse effects , Complement Inactivating Agents/administration & dosage , Complement C5/antagonists & inhibitors
3.
Br J Clin Pharmacol ; 90(5): 1312-1321, 2024 May.
Article in English | MEDLINE | ID: mdl-38373846

ABSTRACT

AIMS: Eculizumab is a monoclonal antibody targeting complement protein C5 used in renal diseases. As recommended dosing regimen leads to unnecessarily high concentrations in some patients, tailored dosing therapeutic drug monitoring was proposed to reduce treatment cost. The objectives of the present work were (i) to investigate the target-mediated elimination of eculizumab and (ii) whether a pharmacokinetic model integrating a nonlinear elimination allows a better prediction of eculizumab concentrations than a linear model. METHODS: We analysed 377 eculizumab serum concentrations from 44 patients treated for atypical haemolytic uraemic syndrome and C3 glomerulopathy with a population pharmacokinetic approach. Critical concentrations (below which a non-log-linear decline of concentration over time is evidenced) were computed to estimate the relevance of the target-mediated elimination. Simulations of dosing regimens were then performed to predict probabilities of target attainment (i.e. trough >100 mg/L). RESULTS: Pharmacokinetics of eculizumab was nonlinear and followed a mixture of first-order (CL = 1.318 mL/day/kg) and Michaelis-Menten elimination (Vmax = 26.07 mg/day, Km = 24.06 mg/L). Volume of distribution (72.39 mL/kg) and clearance were weight-dependent. Critical concentrations (Vmax/CL) ranged from 144.7 to 759.7 mg/L and were inversely related to body weight (P = .013). Nonlinearity was thus noticeable at therapeutic concentrations. Simulations predicted that 1200 mg of eculizumab every 21 days would allow 85% and 76% of patients to maintain a therapeutic exposure, for 50 or 90 kg body weight, respectively. CONCLUSIONS: Our study investigates the nonlinear elimination of eculizumab and discusses the importance of accounting for eculizumab target-mediated elimination in therapeutic drug monitoring.


Subject(s)
Antibodies, Monoclonal, Humanized , Atypical Hemolytic Uremic Syndrome , Drug Monitoring , Models, Biological , Nonlinear Dynamics , Humans , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/administration & dosage , Male , Female , Middle Aged , Adult , Drug Monitoring/methods , Atypical Hemolytic Uremic Syndrome/drug therapy , Aged , Dose-Response Relationship, Drug , Young Adult , Complement Inactivating Agents/pharmacokinetics , Complement Inactivating Agents/administration & dosage , Computer Simulation , Adolescent
4.
Front Immunol ; 12: 785229, 2021.
Article in English | MEDLINE | ID: mdl-34899752

ABSTRACT

The complement system has long been recognized as a potential druggable target for a variety of inflammatory conditions. Very few complement inhibitors have been approved for clinical use, but a great number are in clinical development, nearly all of which systemically inhibit complement. There are benefits of targeting complement inhibition to sites of activation/disease in terms of efficacy and safety, and here we describe P-selectin targeted complement inhibitors, with and without a dual function of directly blocking P-selectin-mediated cell-adhesion. The constructs are characterized in vitro and in murine models of hindlimb ischemia/reperfusion injury and hindlimb transplantation. Both constructs specifically targeted to reperfused hindlimb and provided protection in the hindlimb ischemia/reperfusion injury model. The P-selectin blocking construct was the more efficacious, which correlated with less myeloid cell infiltration, but with similarly reduced levels of complement deposition. The blocking construct also improved tissue perfusion and, unlike the nonblocking construct, inhibited coagulation, raising the possibility of differential application of each construct, such as in thrombotic vs. hemorrhagic conditions. Similar outcomes were obtained with the blocking construct following vascularized composite graft transplantation, and treatment also significantly increased graft survival. This is outcome may be particularly pertinent in the context of vascularized composite allograft transplantation, since reduced ischemia reperfusion injury is linked to a less rigorous alloimmune response that may translate to the requirement of a less aggressive immunosuppressive regime for this normally nonlife-threatening procedure. In summary, we describe a new generation of targeted complement inhibitor with multi-functionality that includes targeting to vascular injury, P-selectin blockade, complement inhibition and anti-thrombotic activity. The constructs described also bound to both mouse and human P-selectin which may facilitate potential translation.


Subject(s)
Cell Adhesion/drug effects , Complement Activation/drug effects , Complement Inactivating Agents/administration & dosage , Hindlimb/blood supply , Hindlimb/transplantation , P-Selectin/antagonists & inhibitors , Receptors, Complement 3b/administration & dosage , Reperfusion Injury/prevention & control , Single-Chain Antibodies/administration & dosage , Vascularized Composite Allotransplantation , Animals , Complement Inactivating Agents/pharmacokinetics , Disease Models, Animal , Fibrinolytic Agents/administration & dosage , Graft Survival/drug effects , Male , Mice, Inbred C57BL , P-Selectin/metabolism , Regional Blood Flow , Reperfusion Injury/immunology , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology , Signal Transduction , Single-Chain Antibodies/pharmacokinetics
5.
Expert Opin Investig Drugs ; 30(5): 483-493, 2021 May.
Article in English | MEDLINE | ID: mdl-33792453

ABSTRACT

INTRODUCTION: Generalized myasthenia gravis (gMG) is an autoimmune disorder in which pathogenic autoantibodies damage the neuromuscular junction, causing disabling or life-threatening muscle weakness. Most treatments nonspecifically inhibit aspects of the immune system, do not directly address the causal mechanisms of tissue damage, and often have side-effect profiles that negatively impact patients. Understanding of the central pathogenic role of the complement cascade in gMG is advancing, and a new complement-targeting treatment is under investigation. AREAS COVERED: We provide an overview of gMG etiology, the complement cascade, current treatments, and the investigational gMG therapy zilucoplan. Zilucoplan is a small, subcutaneously administered, macrocyclic peptide that inhibits cleavage of complement component C5 and the subsequent formation of the membrane attack complex. EXPERT OPINION: In a randomized, double-blind, placebo-controlled, phase 2 clinical trial, zilucoplan demonstrated clinically meaningful complement inhibition in patients with acetylcholine receptor-positive gMG. Zilucoplan, a first-of-its-kind cyclic peptide targeting C5, appears to be a therapeutic option for the treatment of gMG based on available pharmacokinetic/pharmacodynamic data and phase 1 and 2 efficacy, safety, and tolerability data with limited long-term follow-up. Zilucoplan use earlier in the treatment paradigm would be suitable in this population should phase 3 efficacy and safety data be equally favorable.


Subject(s)
Complement Inactivating Agents/pharmacology , Myasthenia Gravis/drug therapy , Animals , Autoantibodies/immunology , Complement C5/antagonists & inhibitors , Complement Inactivating Agents/adverse effects , Complement Inactivating Agents/pharmacokinetics , Humans , Myasthenia Gravis/physiopathology , Randomized Controlled Trials as Topic , Receptors, Cholinergic/immunology
6.
Nat Immunol ; 22(2): 128-139, 2021 02.
Article in English | MEDLINE | ID: mdl-33398182

ABSTRACT

Complement hyperactivation, angiopathic thrombosis and protein-losing enteropathy (CHAPLE disease) is a lethal disease caused by genetic loss of the complement regulatory protein CD55, leading to overactivation of complement and innate immunity together with immunodeficiency due to immunoglobulin wasting in the intestine. We report in vivo human data accumulated using the complement C5 inhibitor eculizumab for the medical treatment of patients with CHAPLE disease. We observed cessation of gastrointestinal pathology together with restoration of normal immunity and metabolism. We found that patients rapidly renormalized immunoglobulin concentrations and other serum proteins as revealed by aptamer profiling, re-established a healthy gut microbiome, discontinued immunoglobulin replacement and other treatments and exhibited catch-up growth. Thus, we show that blockade of C5 by eculizumab effectively re-establishes regulation of the innate immune complement system to substantially reduce the pathophysiological manifestations of CD55 deficiency in humans.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Complement Activation/drug effects , Complement C5/antagonists & inhibitors , Complement Inactivating Agents/therapeutic use , Energy Metabolism/drug effects , Hypoproteinemia/drug therapy , Immunity, Innate/drug effects , Protein-Losing Enteropathies/drug therapy , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Biomarkers/blood , CD55 Antigens/deficiency , CD55 Antigens/genetics , Complement C5/metabolism , Complement Inactivating Agents/adverse effects , Complement Inactivating Agents/pharmacokinetics , Genetic Predisposition to Disease , Humans , Hypoproteinemia/genetics , Hypoproteinemia/immunology , Hypoproteinemia/metabolism , Mutation , Phenotype , Protein-Losing Enteropathies/genetics , Protein-Losing Enteropathies/immunology , Protein-Losing Enteropathies/metabolism , Treatment Outcome
7.
J Allergy Clin Immunol ; 147(4): 1420-1429.e7, 2021 04.
Article in English | MEDLINE | ID: mdl-32926878

ABSTRACT

BACKGROUND: Activation of the classical and lectin pathway of complement may contribute to tissue damage and organ dysfunction of antibody-mediated diseases and ischemia-reperfusion conditions. Complement factors are being considered as targets for therapeutic intervention. OBJECTIVE: We sought to characterize ARGX-117, a humanized inhibitory monoclonal antibody against complement C2. METHODS: The mode-of-action and binding characteristics of ARGX-117 were investigated in detail. Furthermore, its efficacy was analyzed in in vitro complement cytotoxicity assays. Finally, a pharmacokinetic/pharmacodynamic study was conducted in cynomolgus monkeys. RESULTS: Through binding to the Sushi-2 domain of C2, ARGX-117 prevents the formation of the C3 proconvertase and inhibits classical and lectin pathway activation upstream of C3 activation. As ARGX-117 does not inhibit the alternative pathway, it is expected not to affect the antimicrobial activity of this complement pathway. ARGX-117 prevents complement-mediated cytotoxicity in in vitro models for autoimmune hemolytic anemia and antibody-mediated rejection of organ transplants. ARGX-117 exhibits pH- and calcium-dependent target binding and is Fc-engineered to increase affinity at acidic pH to the neonatal Fc receptor, and to reduce effector functions. In cynomolgus monkeys, ARGX-117 dose-dependently reduces free C2 levels and classical pathway activity. A 2-dose regimen of 80 and 20 mg/kg separated by a week, resulted in profound reduction of classical pathway activity lasting for at least 7 weeks. CONCLUSIONS: ARGX-117 is a promising new complement inhibitor that is uniquely positioned to target both the classical and lectin pathways while leaving the alternative pathway intact.


Subject(s)
Antibodies, Monoclonal/pharmacology , Complement C2/antagonists & inhibitors , Complement Inactivating Agents/pharmacology , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacokinetics , Calcium , Complement Activation/drug effects , Complement C2/analysis , Complement C2/metabolism , Complement Inactivating Agents/blood , Complement Inactivating Agents/pharmacokinetics , Epitope Mapping , Female , Humans , Hydrogen-Ion Concentration , Macaca fascicularis , Male
8.
Br J Haematol ; 191(3): 476-485, 2020 11.
Article in English | MEDLINE | ID: mdl-32449174

ABSTRACT

Ravulizumab, a novel long-acting complement component 5 (C5) inhibitor administered every 8 weeks (q8w), was non-inferior to eculizumab for all efficacy outcomes in two randomised, open-label, phase 3 trials in C5 inhibitor-naïve (Study 301) and eculizumab-experienced (Study 302) adult patients with paroxysmal nocturnal haemoglobinuria (PNH). This pre-specified analysis characterised ravulizumab pharmacokinetics (PK), pharmacodynamics (PD; free C5 levels), and PD differences between medications (Study 301, n = 246; Study 302, n = 195). Ravulizumab PK parameters were determined using non-compartmental analysis. Serum free C5 was quantified with a Gyros-based fluorescence assay (ravulizumab) and an electrochemiluminescence ligand-binding assay (eculizumab). Ravulizumab PK parameters were numerically comparable in both studies; the median time to maximum concentrations ranged from 2·3 to 2·8 and 2·3 to 2·6 h in studies 301 and 302, respectively. Ravulizumab steady-state serum concentrations were achieved immediately after the first dose and sustained throughout treatment. For ravulizumab, the mean (SD) post hoc terminal elimination half-life was 49·7 (8·9) days. Serum free C5 concentrations <0·5 µg/ml were achieved after the first ravulizumab dose and sustained throughout treatment in both studies. In a minority of patients, free C5 concentrations <0·5 µg/ml were not consistently achieved with eculizumab in either study. Ravulizumab q8w was more consistent in providing immediate, complete, sustained C5 inhibition than eculizumab every-2-weeks in patients with PNH.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Complement C5/immunology , Complement Inactivating Agents/pharmacokinetics , Hemoglobinuria, Paroxysmal/drug therapy , Hemoglobinuria, Paroxysmal/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Biomarkers/blood , Complement Inactivating Agents/therapeutic use , Female , Hemoglobinuria, Paroxysmal/blood , Hemoglobinuria, Paroxysmal/diagnosis , Humans , Male , Molecular Targeted Therapy , Treatment Outcome
9.
J Pharm Biomed Anal ; 177: 112876, 2020 Jan 05.
Article in English | MEDLINE | ID: mdl-31525575

ABSTRACT

Flavonoids-enriched extract from Scutellaria baicalensis roots (FESR) ameliorated influenza A virus (IAV) induced acute lung injury (ALI) in mice by inhibiting the excessive activation of complement system in vivo. However, FESR had no anti-complementary activity in vitro. In order to reveal the effective materials of FESR for the treatment of IAV-induced ALI, the present research explored the metabolic process of FESR both in nomal and IAV infected mice by the method of UHPLC-ESI-LTQ/MS, as well as the metabolic activating mechanism. The results showed that the inactive flavonoid glycosides of FESR were partly metabolized into anti-complementary aglycones in vivo, mainly including 5,7,4'-trihydroxy-8-methoxy-flavone, norwogonin, baicalein, wogonin, oroxylin A and chrysin. Moreover, compared with the normal mice, IAV-induced ALI mice exhibited more efficient on producing and absorbing these active metabolites, with AUC0-t and Cmax in plasma and concentrations in lungs and intestines markedly elevated in the IAV treated groups (P <  0.05). Interestingly, the intestinal bacteria from IAV-induced ALI mice showed stronger ß-glucuronidase activity and also had higher efficiency on transforming FESR to the flavonoid aglycones. These findings suggested that the anti-complementary aglycones produced by metabolic activation in vivo should be the potential effective materials of FESR against IAV infections, and intestinal bacteria might play an important role on the higher bioavailability of FESR in IAV infected mice. Additionally, the animals under the pathological state are more suitable for the metabolic study of traditional Chinese medicine.


Subject(s)
Acute Lung Injury/drug therapy , Complement Inactivating Agents/pharmacokinetics , Drugs, Chinese Herbal/pharmacokinetics , Flavonoids/pharmacokinetics , Influenza, Human/drug therapy , Scutellaria baicalensis/chemistry , Activation, Metabolic , Acute Lung Injury/metabolism , Acute Lung Injury/virology , Animals , Complement Inactivating Agents/administration & dosage , Complement Inactivating Agents/chemistry , Disease Models, Animal , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/chemistry , Flavonoids/administration & dosage , Gastrointestinal Microbiome/physiology , Glucuronidase/metabolism , Humans , Influenza A virus/immunology , Influenza A virus/pathogenicity , Influenza, Human/metabolism , Influenza, Human/virology , Lung/pathology , Mice , Plant Roots/chemistry , Specific Pathogen-Free Organisms
10.
Am J Kidney Dis ; 74(1): 56-72, 2019 07.
Article in English | MEDLINE | ID: mdl-30851964

ABSTRACT

RATIONALE & OBJECTIVE: Although primary atypical hemolytic uremic syndrome (aHUS) is associated with abnormalities in complement genes and antibodies to complement factor H, the role of complement in secondary aHUS remains debatable. We evaluated the usefulness of an ex vivo test to: (1) detect complement activation within the endothelium in primary and secondary aHUS, (2) differentiate active disease from remission, (3) monitor the effectiveness of eculizumab therapy, and (4) identify relapses during eculizumab dosage tapering and after discontinuation of treatment. STUDY DESIGN: Case series. SETTING & PARTICIPANTS: 121 patients with primary aHUS and 28 with secondary aHUS. Serum samples were collected during acute episodes, following remission, and during eculizumab treatment and were assessed using a serum-induced ex vivo C5b-9 endothelial deposition test. RESULTS: Serum-induced C5b-9 deposition on cultured microvascular endothelium was quantified by calculating the endothelial area covered by C5b-9 staining; values were expressed as percentage of C5b-9 deposits induced by a serum pool from healthy controls. Testing with adenosine diphosphate-activated endothelium demonstrated elevated C5b-9 deposits for all untreated patients with aHUS independent of disease activity, while testing with unstimulated endothelium demonstrated deposits only in active disease. Similar findings were observed in secondary aHUS. Serum-induced C5b-9 deposits on activated and unstimulated endothelium normalized during eculizumab treatment. 96% (22/23) of patients receiving eculizumab at extended 3- or 4-week dosing intervals demonstrated normal C5b-9 deposits on activated endothelium, despite most patients having CH50Eq (serum complement activity) > 20 UEq/mL, indicating that adequate complement control was achieved even with incomplete blockade of circulating C5. During eculizumab dosage tapering or after treatment discontinuation, all patients experiencing relapses versus only 6% (1/17) of those in stable remission had elevated C5b-9 deposits on unstimulated endothelium. LIMITATIONS: The C5b-9 endothelial deposition test can be performed in only specialized laboratories. Findings on eculizumab dosage tapering need to be confirmed with longitudinal monitoring of C5b-9 deposition. CONCLUSIONS: The C5b-9 endothelial deposition assay may represent an advance in our ability to monitor aHUS activity and individualize therapy.


Subject(s)
Antibodies, Monoclonal, Humanized , Atypical Hemolytic Uremic Syndrome , Complement Activation/drug effects , Complement Membrane Attack Complex/analysis , Drug Monitoring/methods , Adult , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Atypical Hemolytic Uremic Syndrome/blood , Atypical Hemolytic Uremic Syndrome/drug therapy , Complement Factor H/analysis , Complement Factor H/genetics , Complement Inactivating Agents/administration & dosage , Complement Inactivating Agents/pharmacokinetics , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Female , Humans , In Vitro Techniques/methods , Male , Reproducibility of Results , Secondary Prevention/methods , Secondary Prevention/statistics & numerical data
11.
Clin Pharmacokinet ; 58(7): 859-874, 2019 07.
Article in English | MEDLINE | ID: mdl-30758736

ABSTRACT

Eculizumab is the first drug approved for the treatment of complement-mediated diseases, and current dosage schedules result in large interindividual drug concentrations. This review provides insight into the pharmacokinetic and pharmacodynamic properties of eculizumab, both for reported on-label (paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, generalized myasthenia gravis) and off-label (hematopoietic stem cell transplantation-associated thrombotic microangiopathy) indications. Furthermore, we discuss the potential of therapeutic drug monitoring to individualize treatment and reduce costs.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Complement Inactivating Agents/pharmacology , Complement Inactivating Agents/pharmacokinetics , Animals , Complement System Proteins , Drug Monitoring , Humans , Precision Medicine
12.
Clin Immunol ; 197: 96-106, 2018 12.
Article in English | MEDLINE | ID: mdl-30217791

ABSTRACT

The central component of the complement cascade, C3, is involved in various biological functions, including opsonization of foreign bodies, clearance of waste material, activation of immune cells, and triggering of pathways controlling development. Given its broad role in immune responses, particularly in phagocytosis and the clearance of microbes, a deficiency in complement C3 in humans is often associated with multiple bacterial infections. Interestingly, an increased susceptibility to infections appears to occur mainly in the first two years of life and then wanes throughout adulthood. In view of the well-established connection between C3 deficiency and infections, therapeutic inhibition of complement at the level of C3 is often considered with caution or disregarded. We therefore set out to investigate the immune and biochemical profile of non-human primates under prolonged treatment with the C3 inhibitor compstatin (Cp40 analog). Cynomolgus monkeys were dosed subcutaneously with Cp40, resulting in systemic inhibition of C3, for 1 week, 2 weeks, or 3 months. Plasma concentrations of both C3 and Cp40 were measured periodically and complete saturation of plasma C3 was confirmed. No differences in hematological, biochemical, or immunological parameters were identified in the blood or tissues of animals treated with Cp40 when compared to those injected with vehicle alone. Further, skin wounds showed no signs of infection in those treated with Cp40. In fact, Cp40 treatment was associated with a trend toward accelerated wound healing when compared with the control group. In addition, a biodistribution study in a rhesus monkey indicated that the distribution of Cp40 in the body is associated with the presence of C3, concentrating in organs that accumulate blood and produce C3. Overall, our data suggest that systemic C3 inhibition in healthy adult non-human primates is not associated with a weakened immune system or susceptibility to infections.


Subject(s)
Complement C3/antagonists & inhibitors , Complement Inactivating Agents/toxicity , Peptides, Cyclic/toxicity , Wound Healing/immunology , Wound Infection/epidemiology , Animals , Complement C3/immunology , Complement C3/metabolism , Complement Inactivating Agents/pharmacokinetics , Macaca fascicularis , Macaca mulatta , Peptides, Cyclic/pharmacokinetics , Time Factors , Tissue Distribution , Wounds and Injuries/immunology
13.
Clin Pharmacol Ther ; 104(4): 655-663, 2018 10.
Article in English | MEDLINE | ID: mdl-29737533

ABSTRACT

Aberrant activation of the classical complement pathway is the common underlying pathophysiology of orphan diseases such as bullous pemphigoid, antibody-mediated rejection of organ transplants, cold agglutinin disease, and warm autoimmune hemolytic anemia. Therapeutic options for these complement-mediated disorders are limited and sutimlimab, a humanized monoclonal antibody directed against complement factor C1s, may be potentially useful for inhibition of the classical complement pathway. A phase I, first-in-human, double-blind, randomized, placebo-controlled, dose-escalation trial of single and multiple doses of sutimlimab or placebo was conducted in 64 volunteers to evaluate safety, tolerability, pharmacokinetic, and pharmacodynamic profiles. Single and multiple infusions of sutimlimab were well tolerated without any safety concerns. sutimlimab exhibited a steep concentration-effect relationship with a Hill coefficient of 2.4, and an IC90 of 15.5 µg/mL. This study establishes the foundation for using sutimlimab as a highly selective inhibitor of the classical complement pathway in different diseases.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Complement C1s/antagonists & inhibitors , Complement Inactivating Agents/administration & dosage , Complement Pathway, Classical/drug effects , Adult , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Austria , Complement C1s/immunology , Complement Inactivating Agents/adverse effects , Complement Inactivating Agents/pharmacokinetics , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Healthy Volunteers , Humans , Infusions, Intravenous , Male , Middle Aged , Treatment Outcome , Young Adult
14.
Clin Pharmacol Ther ; 102(4): 671-678, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28295239

ABSTRACT

Recent studies indicate that eculizumab is often given in excess to atypical hemolytic uremic syndrome (aHUS) patients. Individualization of treatment is thus highly requested; however, data on the pharmacokinetics and pharmacodynamics of eculizumab remain limited. We analyzed 11 patients during induction (weekly), maintenance (2-weekly), and tapering (every 3-8 weeks) phases of treatment. The trough eculizumab levels increased with each additional dose during the induction phase (depending on body weight). During maintenance, high eculizumab concentrations of up to 772 µg/mL were observed. The levels decreased with each following dose during tapering (3- and 4-week intervals); however, three patients maintained target eculizumab levels over long time periods (30-48 weeks). At intervals of 6-8 weeks, target eculizumab levels were no longer attained. Serum samples with eculizumab concentrations ≥50 µg/mL showed adequate complement blockade. Our data provide essential insight for optimization of eculizumab dosing schemes and lessening of therapy burden for the patients and cost of the treatment.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Atypical Hemolytic Uremic Syndrome/drug therapy , Complement Inactivating Agents/administration & dosage , Adult , Antibodies, Monoclonal, Humanized/pharmacokinetics , Child , Child, Preschool , Complement Inactivating Agents/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Infant , Male , Middle Aged , Precision Medicine , Time Factors , Young Adult
15.
J Chromatogr B Analyt Technol Biomed Life Sci ; 1041-1042: 19-26, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27992787

ABSTRACT

Cp40 is a 14-amino acid cyclic analog of the peptidic complement inhibitor compstatin that binds with sub-nanomolar affinity to complement component C3 and has already shown promise in various models of complement-related diseases. The preclinical and clinical development of this compound requires a robust, accurate, and sensitive method for quantitatively monitoring Cp40 in biological samples. In this study, we describe the development and validation of an ultra-high performance liquid chromatography electrospray mass spectrometry method for the quantitation of Cp40 in human and non-human primate (NHP) plasma. Isotope-labeled Cp40 was used as an internal standard, allowing for the accurate and absolute quantitation of Cp40. Labeled and non-labeled Cp40 were extracted from plasma using reversed phase-solid phase extraction, with recovery rates exceeding 80%, indicating minor matrix effects. The triply charged states of Cp40 and isotope-labeled Cp40 were detected at m/z 596.60 and 600.34, respectively, via a Q-TOF mass spectrometer and were used for quantitation. The method was linear in the range of 0.18-3.58µg/mL (r2≥0.99), with precision values below 0.71% in NHP and 0.77% in human plasma. The accuracy of the method ranged from -2.17% to 17.99% in NHP and from -0.26% to 15.75% in human plasma. The method was successfully applied to the quantitation of Cp40 in cynomolgus monkey plasma after an initial intravenous bolus of 2mg/kg followed by repetitive subcutaneous administration at 1mg/kg. The high reproducibility, accuracy, and robustness of the method developed here render it suitable for drug monitoring of Cp40, and potentially other compstatin analogs, in both human and NHP plasma samples during pharmacokinetic and pharmacodynamic studies.


Subject(s)
Chromatography, High Pressure Liquid/methods , Complement Inactivating Agents/blood , Peptides, Cyclic/blood , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Complement Inactivating Agents/chemistry , Complement Inactivating Agents/pharmacokinetics , Female , Humans , Linear Models , Macaca fascicularis , Male , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Solid Phase Extraction
16.
Bioconjug Chem ; 27(10): 2359-2371, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27598771

ABSTRACT

The Ornithodoros moubata Complement Inhibitor (OmCI) binds complement component 5 (C5) with high affinity and, thus, selectively prevents proteolytic activation of the terminal lytic complement pathway. A recombinant version of OmCI (also known as Coversin and rEV576) has proven efficacious in several animal models of complement-mediated diseases and successfully completed a phase Ia clinical trial. Coversin is a small 17 kDa lipocalin protein which has a very short plasma half-life if not bound to C5; therefore, the drug requires frequent dosing. We have improved the pharmacokinetics of Coversin by N-terminal translational conjugation with a 600 residue polypeptide composed of Pro, Ala, and Ser (PAS) residues. To this end, PAS-Coversin as well as the unmodified Coversin were functionally expressed in the cytoplasm of E. coli and purified to homogeneity. Both versions showed identical affinity to human C5, as determined by surface plasmon resonance measurements, and revealed similar complement inhibitory activity, as measured in ELISAs with human serum. In line with the PEG-like biophysical properties, PASylation dramatically prolonged the plasma half-life of uncomplexed Coversin by a factor ≥50 in mice. In a clinically relevant in vitro model of the complement-mediated disease paroxysmal nocturnal hemoglobinuria (PNH) both versions of Coversin effectively reduced erythrocyte lysis. Unexpectedly, while the IC50 values were comparable, PAS-Coversin reached a substantially lower plateau of residual lysis at saturating inhibitor concentrations. Taken together, our data demonstrate two clinically relevant improvements of PASylated Coversin: markedly increased plasma half-life and considerably reduced background hemolysis of erythrocytes with PNH-induced phenotype.


Subject(s)
Complement Inactivating Agents/chemistry , Complement Inactivating Agents/pharmacology , Animals , Circular Dichroism , Complement C5/antagonists & inhibitors , Complement Inactivating Agents/pharmacokinetics , Erythrocytes/drug effects , Female , Hemoglobinuria, Paroxysmal/drug therapy , Hemolysis/drug effects , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Engineering/methods , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Surface Plasmon Resonance
17.
Exp Eye Res ; 145: 352-358, 2016 04.
Article in English | MEDLINE | ID: mdl-26919788

ABSTRACT

Uncontrolled activation of complement and upregulation of vascular endothelial growth factor (VEGF) play fundamental roles in age-related macular degeneration (AMD). However, most drugs used to treat AMD focus on a single target, and the percentage of effectively treated patients in clinical practice needs to be improved. Therefore, novel AMD treatment approaches are needed. IBI302 is a novel bispecific decoy receptor fusion protein designed with both a VEGF inhibition domain and a complement cascade inhibition domain, which are connected by the Fc region of human immunoglobulin. In this study, we systematically evaluated the binding affinity between IBI302 and VEGF isoforms and complement proteins by using surface plasmon resonance (SPR) technology. Anti-VEGF blockers (aflibercept and bevacizumab) and complement receptor 1 were used as references. The SPR assay results indicated that IBI302 could bind different VEGF isoforms and complement proteins with high affinity. The biological activity of IBI302 was also studied. IBI302 showed an inhibitory effect on human primary umbilical vein endothelial cell proliferation and the activation of complement pathways in vitro. Finally, the pharmacokinetic (PK) properties of IBI302 were evaluated in rhesus monkeys. The PK results showed that after a 0.5 mg/eye intravitreal dosage, IBI302 became rapidly distributed from the vitreous humor into targeted tissues and remained active over 504 h. Overall, the favorable anti-angiogenic and anti-complement effects of IBI302 along with the good PK profiles in rhesus monkeys support the selection and development of IBI302 as a promising candidate for AMD treatment.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Complement Activation/drug effects , Complement Inactivating Agents/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Macular Degeneration/drug therapy , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacokinetics , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Cell Proliferation/drug effects , Complement Inactivating Agents/pharmacokinetics , Complement System Proteins/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Intravitreal Injections , Macaca mulatta , Recombinant Fusion Proteins/pharmacokinetics , Vitreous Body/metabolism
18.
Kidney Int ; 89(3): 701-11, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26880462

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is caused by alternative complement pathway dysregulation, leading to systemic thrombotic microangiopathy (TMA) and severe end-organ damage. Based on 2 prospective studies in mostly adults and retrospective data in children, eculizumab, a terminal complement inhibitor, is approved for aHUS treatment. Here we prospectively evaluated efficacy and safety of weight-based dosing of eculizumab in eligible pediatric patients with aHUS in an open-label phase II study. The primary end point was complete TMA response by 26 weeks. Twenty-two patients (aged 5 months-17 years) were treated; 16 were newly diagnosed, 12 had no prior plasma exchange/infusion during current TMA symptomatology, 11 received baseline dialysis and 2 had prior renal transplants. By week 26, 14 achieved a complete TMA response, 18 achieved hematologic normalization, and 16 had 25% or better improvement in serum creatinine. Plasma exchange/infusion was discontinued in all, and 9 of the 11 patients who required dialysis at baseline discontinued, whereas none initiated new dialysis. Eculizumab was well tolerated; no deaths or meningococcal infections occurred. Bone marrow failure, wrist fracture, and acute respiratory failure were reported as unrelated severe adverse events. Thus, our findings establish the efficacy and safety of eculizumab for pediatric patients with aHUS and are consistent with proposed immediate eculizumab initiation following diagnosis in children.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/drug therapy , Complement Activation/drug effects , Complement Inactivating Agents/therapeutic use , Adolescent , Age Factors , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Atypical Hemolytic Uremic Syndrome/diagnosis , Atypical Hemolytic Uremic Syndrome/immunology , Australia , Child , Child, Preschool , Complement Inactivating Agents/adverse effects , Complement Inactivating Agents/pharmacokinetics , Europe , Female , Humans , Infant , Male , North America , Plasma Exchange , Prospective Studies , Renal Dialysis , Time Factors , Treatment Outcome
19.
ChemMedChem ; 9(10): 2223-6, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25056114

ABSTRACT

The compstatin family of complement inhibitors has shown promise in various immuno-inflammatory disorders. Although recent analogues show beneficial pharmacokinetics, further extension of the plasma half-life is expected to benefit systemic application of these peptidic inhibitors. We therefore synthesized conjugates of compstatin analogues and albumin-binding molecules (ABM) to increase circulatory residence. Equilibrium dialysis in complement-depleted serum showed a marked increase in plasma protein binding from <8 % to >99 % for a resulting chimera (ABM2-Cp20). Further analysis confirmed interaction with albumin from different species, primarily via site II. Importantly, ABM2-Cp20 bound 20-fold stronger to its target protein C3b (KD =150 pM) than the parent peptide. Kinetic and in silico analysis suggested that ABM2 occupies a secondary site on C3b and improves the dissociation rate via additional contacts. Addition of an ABM modifier thereby not only improved plasma protein binding but also produced the most potent compstatin analogue to date with potential implications for the treatment of systemic complement-related diseases.


Subject(s)
Albumins/chemistry , Complement Inactivating Agents/pharmacology , Peptides, Cyclic/pharmacology , Amino Acid Sequence , Complement Inactivating Agents/pharmacokinetics , Models, Molecular , Molecular Sequence Data , Peptides, Cyclic/pharmacokinetics , Protein Binding
20.
Transfusion ; 49(9): 1987-95, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19497056

ABSTRACT

BACKGROUND: Hereditary angioedema (HAE) caused by functional deficiency of C1-inhibitor (C1-INH) is a rare disease that manifests with recurrent spontaneous nonallergic edema of the subcutaneous tissues and mucous membranes. In cases of laryngeal edema that are not treated immediately, HAE is associated with high mortality rates. Attenuated androgens (e.g., danazol) are usually administered for prophylaxis, but associated side effects may limit their use. This study investigated the efficacy, safety, and quality of life (QoL) associated with a pasteurized plasma-derived C1-inhibitor (pC1-INH) concentrate for individual replacement therapy (IRT) in patients with severe HAE suffering from frequent attacks who were intolerant or not responding to danazol. STUDY DESIGN AND METHODS: Twenty-two patients with severe HAE and danazol incompatibility or insufficient efficacy of danazol were recruited. Intraindividual comparisons of efficacy, safety, and QoL with pC1-INH concentrate IRT versus danazol treatment were made using retrospective and prospective patient data. Pharmacokinetic data were collected for 15 of the 22 patients. RESULTS: In patients receiving pC1-INH regularly, the median number of attacks per year decreased significantly compared to danazol prophylaxis (p < 0.001), and the 24 laryngeal edema episodes per year ceased. Superior efficacy of pC1-INH was found for all QoL variables (e.g., general condition, social activities). No transmission of human immunodeficiency virus or hepatitis A, B, or C was observed. CONCLUSION: In patients with severe HAE who experience severe side effects and/or lack of efficacy of danazol prophylaxis, very early substitution with pC1-INH can completely abolish the incidence of potentially fatal laryngeal edema and can reduce the incidence of acute attacks.


Subject(s)
Angioedemas, Hereditary/drug therapy , Angioedemas, Hereditary/prevention & control , Complement C1 Inhibitor Protein/therapeutic use , Complement Inactivating Agents/pharmacokinetics , Complement Inactivating Agents/therapeutic use , Danazol/therapeutic use , Estrogen Antagonists/therapeutic use , Adult , Complement C1 Inhibitor Protein/pharmacokinetics , Drug Resistance/physiology , Female , Humans , Male , Middle Aged , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...