scholarly journals Targeting the Complement Serine Protease MASP-2 as a Therapeutic Strategy for Coronavirus Infections

Viruses ◽  
2021 ◽  
Vol 13 (2) ◽  
pp. 312
Author(s):  
Ben M. Flude ◽  
Giulio Nannetti ◽  
Paige Mitchell ◽  
Nina Compton ◽  
Chloe Richards ◽  
...  

MASP-2, mannose-binding protein-associated serine protease 2, is a key enzyme in the lectin pathway of complement activation. Hyperactivation of this protein by human coronaviruses SARS-CoV, MERS-CoV and SARS-CoV-2 has been found to contribute to aberrant complement activation in patients, leading to aggravated lung injury with potentially fatal consequences. This hyperactivation is triggered in the lungs through a conserved, direct interaction between MASP-2 and coronavirus nucleocapsid (N) proteins. Blocking this interaction with monoclonal antibodies and interfering directly with the catalytic activity of MASP-2, have been found to alleviate coronavirus-induced lung injury both in vitro and in vivo. In this study, a virtual library of 8736 licensed drugs and clinical agents has been screened in silico according to two parallel strategies. The first strategy aims at identifying direct inhibitors of MASP-2 catalytic activity, while the second strategy focusses on finding protein-protein interaction inhibitors (PPIs) of MASP-2 and coronaviral N proteins. Such agents could represent promising support treatment options to prevent lung injury and reduce mortality rates of infections caused by both present and future-emerging coronaviruses. Forty-six drug repurposing candidates were purchased and, for the ones selected as potential direct inhibitors of MASP-2, a preliminary in vitro assay was conducted to assess their interference with the lectin pathway of complement activation. Some of the tested agents displayed a dose-response inhibitory activity of the lectin pathway, potentially providing the basis for a viable support strategy to prevent the severe complications of coronavirus infections.

Author(s):  
Ting Gao ◽  
Mingdong Hu ◽  
Xiaopeng Zhang ◽  
Hongzhen Li ◽  
Lin Zhu ◽  
...  

AbstractAn excessive immune response contributes to SARS-CoV, MERS-CoV and SARS-CoV-2 pathogenesis and lethality, but the mechanism remains unclear. In this study, the N proteins of SARS-CoV, MERS-CoV and SARS-CoV-2 were found to bind to MASP-2, the key serine protease in the lectin pathway of complement activation, resulting in aberrant complement activation and aggravated inflammatory lung injury. Either blocking the N protein:MASP-2 interaction or suppressing complement activation can significantly alleviate N protein-induced complement hyper-activation and lung injury in vitro and in vivo. Complement hyper-activation was also observed in COVID-19 patients, and a promising suppressive effect was observed when the deteriorating patients were treated with anti-C5a monoclonal antibody. Complement suppression may represent a common therapeutic approach for pneumonia induced by these highly pathogenic coronaviruses.One Sentence SummaryThe lectin pathway of complement activation is a promising target for the treatment of highly pathogenic coronavirus induced pneumonia.


Author(s):  
Min Wei ◽  
Wei-yi Guo ◽  
Bo-yang Xu ◽  
Su-fang Shi ◽  
Li-jun Liu ◽  
...  

Background and objectives: IgA nephropathy is the most common primary glomerulonephritis worldwide. Previous research demonstrated that collectin11, an initiator of complement lectin pathway, was involved in both acute kidney injury and chronic tubulointerstitial fibrosis. Here, we investigated the potential role of collectin11 in the pathogenesis of IgA nephropathy. Design, setting, participants, and measurements: The deposition of collectin11 and other complement proteins was detected in glomeruli of 60 participants with IgA nephropathy by immunofluorescence. In vitro, human mesangial cells were treated with IgA1-containing immune complexes derived from participants with IgA nephropathy. Then, the expression of collectin11 in mesangial cells was examined by RT-qPCR and immunofluorescence. The codeposition of collctin11 with IgA1 or C3 on mesangial cells was detected by immunofluorescence and proximity ligation assays. Results: 37% participants with IgA nephropathy (22/60) showed codeposition of collectin11 with IgA in the glomerular mesangium. Using an injury model of mesangial cells, we demonstrated that IgA1-immune complexes derived from participants with IgA nephropathy increased the secretion of collectin11 in mesangial cells with the subsequent deposition of collectin11 on the cell surface via the interaction with deposited IgA1-immune complexes. In vitro, we found that collectin11 bound to IgA1-immune complexes in a dose-dependent but calcium-independent manner. Furthermore, deposited collectin11 initiated the activation of complement and accelerated the deposition of C3 on mesangial cells. Conclusions: In situ-produced collectin11 by mesangial cells might play an essential role in kidney injury in a subset of patients with IgA nephropathy through the induction of complement activation.


1988 ◽  
Vol 254 (2) ◽  
pp. H258-H264
Author(s):  
L. H. Bruner ◽  
K. J. Johnson ◽  
G. O. Till ◽  
R. A. Roth

Monocrotaline pyrrole (MCTP) causes pulmonary vascular injury, pulmonary hypertension, and right ventricular hypertrophy in rats. The mechanisms by which MCTP causes lung injury are not known. After treatment with a moderate dose of MCTP, several days pass before major lung injury is detected, thus suggesting that the damage is caused indirectly. Since activation of the complement system can cause lung injury, it was of interest to test whether complement activation may be important in lung injury due to MCTP. Accordingly, rats were given a single dose of MCTP (3.5 mg/kg iv), and serum hemolytic complement activity was measured at several times after rats were treated. Neutrophil aggregometry also was used to determine whether complement activation products could be detected in serum after MCTP was given in vivo. The effect of complement depletion on MCTP-induced pulmonary injury was tested by cotreating rats with purified cobra venom factor and MCTP. MCTP treatment did not cause detectable complement activation in vivo, and complement depletion did not protect rats from lung injury. The direct effect of MCTP on serum complement also was tested by exposing fresh rat serum to MCTP in vitro and measuring serum complement activity. MCTP decreased serum hemolytic complement activity in vitro, but it did not interfere with subsequent zymosan-induced activation of complement. These results suggest that complement does not play a role in the development of major lung injury that occurs several days after treatment of rats with MCTP.


2011 ◽  
Vol 300 (4) ◽  
pp. F932-F940 ◽  
Author(s):  
Pieter van der Pol ◽  
Anja Roos ◽  
Stefan P. Berger ◽  
Mohamed R. Daha ◽  
Cees van Kooten

Ischemia-reperfusion injury (IRI) has a major impact on graft survival after transplantation. Renal proximal tubular epithelial cells (PTEC) located at the corticomedullary zone are relatively susceptible to IRI and have been identified as one of the main targets of complement activation. Studies in mice have shown an important role for the alternative pathway of complement activation in renal IRI. However, it is unclear whether experimental data obtained in mice can be extrapolated to humans. Therefore, we developed an in vitro model to induce hypoxia-reoxygenation in human and mouse PTEC and studied the role of the different pathways of complement activation. Exposure of human PTEC to hypoxia followed by reoxygenation in human serum resulted in extensive complement activation. Inhibition studies using different complement inhibitors revealed no involvement of the alternative or lectin pathway of complement activation by hypoxic human PTEC. In contrast, complement activation by hypoxic murine PTEC was shown to be exclusively dependent on the alternative pathway. Hypoxic human PTEC induced classic pathway activation, supported by studies in C1q-depleted serum and the use of blocking antibodies to C1q. The activation of the classic pathway was mediated by IgM through interaction with modified phosphomonoesters exposed on hypoxic PTEC. Studies with different human sera showed a strong correlation between IgM binding to hypoxic human PTEC and the degree of complement activation. These results demonstrate important species-specific differences in complement activation by hypoxic PTEC and provide clues for directed complement inhibition strategies in the treatment and prevention of IRI in the human kidney.


2020 ◽  
Vol 2020 ◽  
pp. 1-12 ◽  
Author(s):  
Yun-peng Wang ◽  
Yu Guo ◽  
Ping-shan Wen ◽  
Zhen-zhen Zhao ◽  
Jian Xie ◽  
...  

Xuebijing injection is a Chinese herb compound to treat sepsis in China, but it contains many different kinds of components, and each component may have different effects in treating sepsis. The present study was performed to investigate the effect of three ingredients of Xuebijing, safflor yellow A (SYA), hydroxysafflor yellow A (HSYA), and anhydrosafflor yellow B (AHSYB), in lipopolysaccharide- (LPS-) induced acute lung injury (ALI). LPS (10 mg/kg) was injected intratracheally to induce acute lung injury in mice, which were then treated with SYA, HSYA, and AHSYB. The blood, bronchoalveolar lavage fluid (BALF), and lung tissues were collected to detect degree of lung injury, level of inflammation, and neutrophil extracellular traps (NETs). In vitro experiments were performed using HL-60 cells stimulated with phorbol myristate acetate (PMA). Lung injury induced by LPS was alleviated by SYA, HSYA, and AHSYB as demonstrated by the histopathologic test. The three components inhibit LPS-induced elevation of the levels of inflammatory factors and wet-to-dry weight ratio as well as the amount of protein and cells in the BALF. They also induced a remarkably less overlay of myeloperoxidase (MPO) and histone in the immunofluorescence assay and reduced level of MPO-DNA complex in plasma. The in vitro assay showed a similar trend that the three components inhibited PMA-induced NET release in neutrophil-like HL-60 cells. Western blot demonstrated that phosphorylation of c-rapidly accelerated fibrosarcoma (c-Raf), mitogen-activated protein kinase ERK kinase (MEK), and extracellular signal-regulated kinase (ERK) in the lungs of LPS-challenged mice, and PMA-treated HL-60 cells were all significantly reduced by SYA, HSYA, and AHSYB. Therefore, our data demonstrated that three components of XBJ, including SYA, HSYA, and AHSYB, showed a protective effect against LPS-induced lung injury and NET release.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 26-27
Author(s):  
Jeremy Freeman ◽  
Jason Cummings ◽  
Marither Chuidian ◽  
Thomas Dudler

Introduction Narsoplimab (OMS721) is a fully human monoclonal antibody that binds to and inhibits mannan-binding lectin-associated serine protease 2 (MASP-2). MASP-2 is the key enzyme responsible for activation of the lectin pathway of the complement system. The lectin pathway is activated when plasma is exposed to molecular patterns present on microbes and injured host cell surfaces, initiating a proteolytic cascade that results in the activation of complement components C4, C2, C3 and C5 to yield multiple biologically active products. Inhibiting MASP-2 activity prevents lectin-mediated generation of complement activation products, thereby reducing inflammation and tissue injury. Narsoplimab is the first lectin pathway-specific complement inhibitor in clinical development. In published clinical studies and case reports, improvement has been observed in narsoplimab-treated patients with hematopoietic stem cell transplant-associated thrombotic microangiopathy, immunoglobulin A (IgA) nephropathy and atypical hemolytic uremic syndrome. Here we describe the development of pharmacodynamic (PD) assays to quantify MASP-2 inhibition ex vivo and their use to study duration of action and to establish the pharmacokinetic (PK)/PD relationship of narsoplimab in monkeys and humans. Methods PD assays to measure lectin pathway activity, a measure of MASP-2 activity, in minimally diluted (90%) serum from human and monkey were developed using immobilized mannan as a lectin pathway-specific complement activator with C4b as the activation endpoint. ELISA and flow cytometry assay platforms were employed to assess the PD response of monkeys or normal healthy human volunteers administered narsoplimab, respectively. The PD response was used to characterize the time-course and dose-response of lectin pathway inhibition by narsoplimab. Drug concentration data obtained from matched serum samples were used to estimate the ex vivo EC50 of lectin pathway inhibition in monkeys and humans. Lectin pathway-specific complement activation assays applicable to minimally diluted serum samples were developed by evaluating the time-course of lectin-induced complement activation response under varying reaction temperatures and selecting optimized test conditions within the dynamic range of the assay platform. The test methods were used to characterize monkey and human lectin pathway inhibition profiles of narsoplimab in vitro, and the flow cytometric method was validated for analysis of human serum samples. Results The PD assays were used to characterize the PD response of monkeys administered a single intravenous (IV) bolus injection of narsoplimab at 0.05, 0.15, 0.5, 1.5 or 5 mg/kg, and of normal human healthy volunteers administered a single IV infusion of the drug at 0.25, 0.625 or 2 mg/kg, or 6 weekly IV infusions at 2 or 4 mg/kg. A clear, dose-dependent initial PD response was observed in monkeys in the dose range of 0.5 mg/kg to 5 mg/kg, which subsequently declined over time. In humans, a minimal PD response was observed at 0.025 mg/kg. A near maximal initial PD response was seen at 0.675 mg/kg or higher, followed by a decline over time. In the 6-week repeat-dose study, once-weekly dosing of narsoplimab at 2 mg/kg resulted in a mean trough PD response of ~55% lectin pathway inhibition, which increased to an ~80% mean trough response with once-weekly dosing of 4 mg/kg. PK sample analysis revealed a clear PK/PD relationship in monkeys and humans. Modeling of the PK/PD relationship in monkeys indicated an EC50 value of ~7 µg/mL (~ 46 nM), which is comparable to the IC50 value of lectin pathway inhibition in vitro (33 nM). In humans, modeling of the PK/PD relationship data indicated an EC50 value of ~500 ng/mL (3.3 nM), again very similar to the IC50 value of lectin pathway inhibition in vitro (3.4 nM). Conclusion We successfully developed PD assays to monitor blockade of lectin pathway activation in monkeys and humans administered narsoplimab. Using a validated PD assay, we demonstrate that a high level of lectin pathway blockade can be maintained in normal human healthy volunteers by once-weekly administration of narsoplimab at 4 mg/kg IV. We also demonstrate a consistent PK/PD relationship across the study population, indicating that narsoplimab concentration ranges can be used to target the desired level of lectin pathway inhibition. Figure 1 Disclosures Freeman: Omeros Corporation: Current Employment. Cummings:Omeros Corporation: Current Employment. Chuidian:Omeros Corporation: Current Employment. Dudler:Omeros Corporation: Current Employment.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 354-354
Author(s):  
Saskia van der Velden ◽  
Thijs L.J. van Osch ◽  
Amina Seghier ◽  
Arthur Bentlage ◽  
Juk Yee Mok ◽  
...  

Abstract Transfusion-related acute lung injury (TRALI) is a leading cause of blood transfusion-related fatalities without available therapies. The pulmonary endothelium is damaged in TRALI, through incompletely understood pathogenic mechanisms, resulting in pulmonary edema. Generally, anti-leukocyte antibodies or biological response modifiers in the transfusion product, in combination with predisposing risk factors in the transfused recipient (e.g. inflammation), are responsible for initiation of TRALI. Remarkably, not all anti-leukocyte antibodies cause TRALI. In a previous in vitro study, we identified increased Fc-mediated complement activation to be a key feature of murine TRALI-inducing antibody 34-1-2S (anti-H-2K d) compared to TRALI-resistant antibody SF1.1.10 (anti-H-2K d) (Zeeuw van der Laan et al, Blood Adv 2020). In the current study, we further explored antibody-mediated TRALI mechanisms in vivo using our previously established TRALI mouse model in which mice are pre-depleted of protective CD4+ T cells and primed with LPS, followed by infusion of antibody 34-1-2S (Kapur et al, Blood 2017, Blood Adv 2018, Blood 2019). A key read-out for TRALI was the lung wet/dry weight ratio (Lung W/D, measure for pulmonary edema). We found that in vitro antibody-mediated complement activation was associated with in vivo antibody-mediated TRALI. 34-1-2S caused severe TRALI (complement activation: +++, Lung W/D: 7.4 ± 0.21), while SF1.1.10 did not cause TRALI (complement activation: +, Lung W/D: 4.68 ± 0.16). Furthermore, Fc-deglycosylated 34-1-2S still caused significant TRALI (complement activation: ++, in vivo Lung W/D: 5.16 ± 0.52). TRALI development was fully Fc-dependent as 34-1-2S-Fab did not cause any TRALI (no complement activation, Lung W/D: 4.35 ± 0.18). Importantly, we found significantly increased levels of complement C1q-C4 complexes in plasma samples of TRALI patients (n=46) compared to healthy controls (n=25): 6.49 ± 5.27 vs 3.84 ± 2.27 AU/ml, respectively, P: 0.0005,***. Similarly, C5a levels were significantly elevated in plasma samples of TRALI patients (n=53) compared to healthy controls (n=30): 2.52 ± 2.17 vs 1.37 ± 1.08 ng/ml, respectively, P: 0.0006,***. To further dissect the effect of the Fc-part of 34-1-2S in TRALI-induction, we aim to investigate the contribution of Fc-mediated complement activation vs Fc-receptor interaction. We therefore successfully generated chimeric variants of 34-1-2S with a humanized IgG1 Fc-domain containing mutations making them functionally complement dead (K322A), Fc-receptor dead (dG236) or both complement and Fc-receptor dead (PG LALA). We are currently investigating the effects of these variants on in vivo TRALI induction. We next investigated how the in vivo murine TRALI reaction was related to numbers of macrophages, monocytes and neutrophils in blood and lungs. We found that 34-1-2S-TRALI was associated with significantly decreased levels of macrophages in the lungs and increased levels in blood, compared to mice infused with PBS, SF1.1.10 or 34-1-2S-Fab, suggesting that Fc-mediated complement activation and TRALI induction is related to macrophage trafficking from lungs to blood. We did not observe any significant differences between blood and lung neutrophil levels of 34-1-2S-TRALI mice compared to SF1.1.10 TRALI-resistant mice. We hypothesized that in TRALI neutrophils undergo formation of neutrophil extracellular traps (NETs) induced by complement. We observed that C5a enabled potent neutrophil-chemotaxis in vitro (P: 0.0048,**). In addition, using direct immunofluorescence staining of extracellular DNA and Citrullinated histone H3, we observed that both LPS and C5a on their own could induce NET formation in vitro, which was synergistically increased with a combination of both LPS and C5a (P: 0.0417,*), as occurring during TRALI. Furthermore, we found increased levels of NETs to be present in plasma samples of TRALI patients (n=53) compared to healthy controls (n=30): 1.64 ± 0.97 vs 0.80 ± 0.34 MPO-DNA OD, respectively, P: 0.0002,***. Finally, we targeted the C5a-receptor (C5aR) using a C5aR-antagonist in our TRALI mouse model. Surprisingly, this did not prevent but even worsened TRALI (P: 0.0398,*), with elevated levels of blood monocytes and macrophages. This suggests that an approach directly targeting complement components may be a more promising therapeutic strategy to explore in combatting TRALI. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1453-1453
Author(s):  
Ellinor I.B. Peerschke ◽  
Sandip Panicker ◽  
Alexa M. Sughroue ◽  
James B. Bussel

Abstract Background: Immune thrombocytopenic purpura (ITP) is an autoimmune disorder in which antiplatelet antibodies mediate accelerated platelet clearance from circulation and also inhibit platelet production, resulting in thrombocytopenia. Activation of the classical pathway (CP) of complement is associated with a variety of immune disorders involving the presence of autoantibodies. The role of the complement system in ITP is poorly understood. Methods: Plasma samples (0.32% sodium citrate) from patients with chronic ITP (n=55) were evaluated for their ability to activate the CP of complement. The 55 patients consisted primarily of adult but also of pediatric patients with ITP, undergoing various treatment regimens. The most common included IVIG, rituximab, and especially thrombopoietic agents (eltrombopag, romiplostim). Almost all patients included in the analysis had chronic ITP, defined as ITP lasting > 12 months. The complement activating capacity (CAC) of patient plasma was evaluated with a previously described in vitro assay ( Peerschke et al., Brit J Haematol, 2009) that measures complement activation on immobilized, fixed heterologous platelets using an ELISA approach with monoclonal antibodies to C1q, C4d, iC3b, and C5b-9. CAC represents assay optical density readings normalized to reference normal plasma pool. A CAC of >1.5 was considered indicative of enhanced complement activation, based on reference ranges established for plasma from healthy volunteers. Patient CAC values were correlated with platelet count. The ability of TNT003 to block in vitro complement activation was assessed relative to an isotype matched control. TNT003 is a mouse monoclonal antibody (IgG2a) that targets the CP-serine protease C1s. Results: A statistically significant (p=0.042) inverse correlation was noted between C4d deposition and platelet count in the 55 ITP patient samples tested. Heightened classical complement pathway activation was demonstrated in 7 of 55 patients (~13%) with ITP as evidenced by increased C4d deposition. 6 of the 7 patients with increased C4d deposition had platelet counts <100k/mcL, and 5 patients had platelet counts <50K/mcL. There was a non-significant trend for higher C4d levels on platelets and lower AIPF (absolute immature platelet fraction, equivalent to platelet reticulocytes). TNT003 (100 mcg/ml) inhibited C4 activation by 44 + 43% in ITP plasma in vitro. Inhibition of downstream complement activation, iC3b and C5b-9 deposition, was 72% + 17 % and 82% + 14% (mean + S.D.), respectively. Similar results were obtained using 10 mM EDTA, a known inhibitor of complement activation. Conclusions: The heterogeneity of patient responses to different treatment modalities in ITP support the concept of different immune mechanisms contributing to thrombocytopenia. Our data demonstrate classical complement pathway activation in a subgroup of patients with ITP, and further present the first evidence of CP complement inhibition by a novel C1s inhibitor in this setting. Failure to completely block C4 activation in ITP plasma in vitro by either of TNT003 or EDTA, suggests the presence of preformed, circulating C4d containing complement complexes in patient plasma. The ability of TNT003 to more completely inhibit C3 activation and C5b-9 assembly downstream of C4 in the in vitro assay system is consistent with direct activation and inhibition of complement at the platelet surface. Thus, TNT003 may mitigate enhanced platelet clearance by RES via inhibition of complement mediated platelet opsonization by C3b and platelet lysis by C5b-9. Further studies are required to evaluate the impact of TNT003 on thrombocytopenia in ITP. Disclosures Peerschke: True North Therapeutics: Research Support Other. Panicker:True North Therapeutics: Employment. Bussel:True North Therapeutics: Research Support Other.


2000 ◽  
Vol 49 (1-2) ◽  
pp. 79 ◽  
Author(s):  
M.R. Dahl ◽  
S. Thiel ◽  
A.C. Willis ◽  
T. Vorup-Jensen ◽  
T. Christensen ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document