scholarly journals Homodimeric Minimal Factor H: In Vivo Tracking and Extended Dosing Studies in Factor H Deficient Mice

2021 ◽  
Vol 12 ◽  
Author(s):  
Ola Kamala ◽  
Talat H. Malik ◽  
Thomas M. Hallam ◽  
Thomas E. Cox ◽  
Yi Yang ◽  
...  

C3 glomerulopathy (C3G) is associated with dysregulation of the alternative pathway (AP) of complement and treatment options remain inadequate. Factor H (FH) is a potent regulator of the AP. An in-depth analysis of FH-related protein dimerised minimal (mini)-FH constructs has recently been published. This analysis showed that addition of a dimerisation module to mini-FH not only increased serum half-life but also improved complement regulatory function, thus providing a potential treatment option for C3G. Herein, we describe the production of a murine version of homodimeric mini-FH [mHDM-FH (mFH1–5^18–20^R1–2)], developed to reduce the risk of anti-drug antibody formation during long-term experiments in murine models of C3G and other complement-driven pathologies. Our analysis of mHDM-FH indicates that it binds with higher affinity and avidity to WT mC3b when compared to mouse (m)FH (mHDM-FH KD=505 nM; mFH KD=1370 nM) analogous to what we observed with the respective human proteins. The improved binding avidity resulted in enhanced complement regulatory function in haemolytic assays. Extended interval dosing studies in CFH-/- mice (5mg/kg every 72hrs) were partially effective and bio-distribution analysis in CFH-/- mice, through in vivo imaging technologies, demonstrates that mHDM-FH is preferentially deposited and remains fixed in the kidneys (and liver) for up to 4 days. Extended dosing using an AAV- human HDM-FH (hHDM-FH) construct achieved complete normalisation of C3 levels in CFH-/- mice for 3 months and was associated with a significant reduction in glomerular C3 staining. Our data demonstrate the ability of gene therapy delivery of mini-FH constructs to enhance complement regulation in vivo and support the application of this approach as a novel treatment strategy in diseases such as C3G.

2018 ◽  
Vol 29 (6) ◽  
pp. 1649-1661 ◽  
Author(s):  
Yi Yang ◽  
Harriet Denton ◽  
Owen R. Davies ◽  
Kate Smith-Jackson ◽  
Heather Kerr ◽  
...  

Background C3 glomerulopathy (C3G) is associated with dysregulation of the alternative pathway of complement activation, and treatment options for C3G remain limited. Complement factor H (FH) is a potent regulator of the alternative pathway and might offer a solution, but the mass and complexity of FH makes generation of full-length FH far from trivial. We previously generated a mini-FH construct, with FH short consensus repeats 1–5 linked to repeats 18–20 (FH1–5^18–20), that was effective in experimental C3G. However, the serum t1/2 of FH1–5^18–20 was significantly shorter than that of serum-purified FH.Methods We introduced the oligomerization domain of human FH-related protein 1 (denoted by R1–2) at the carboxy or amino terminus of human FH1–5^18–20 to generate two homodimeric mini-FH constructs (FHR1–2^1–5^18–20 and FH1–5^18–20^R1–2, respectively) in Chinese hamster ovary cells and tested these constructs using binding, fluid-phase, and erythrocyte lysis assays, followed by experiments in FH-deficient Cfh−/− mice.Results FHR1–2^1–5^18–20 and FH1–5^18–20^R1–2 homodimerized in solution and displayed avid binding profiles on clustered C3b surfaces, particularly FHR1–2^1–5^18–20. Each construct was >10-fold more effective than FH at inhibiting cell surface complement activity in vitro and restricted glomerular basement membrane C3 deposition in vivo significantly better than FH or FH1–5^18–20. FH1–5^18–20^R1–2 had a C3 breakdown fragment binding profile similar to that of FH, a >5-fold increase in serum t1/2 compared with that of FH1–5^18–20, and significantly better retention in the kidney than FH or FH1–5^18–20.Conclusions FH1–5^18–20^R1–2 may have utility as a treatment option for C3G or other complement-mediated diseases.


2017 ◽  
Vol 45 (12) ◽  
pp. 2824-2834 ◽  
Author(s):  
Anne C.T. Vrancken ◽  
Gerjon Hannink ◽  
Wojciech Madej ◽  
Nico Verdonschot ◽  
Tony G. van Tienen ◽  
...  

Background: Injury or loss of the meniscus generally leads to degenerative osteoarthritic changes in the knee joint. However, the treatment options for symptomatic patients with total meniscectomy are limited. Therefore, we developed a novel, anatomically shaped, total meniscal implant made of polycarbonate urethane. Purpose: To evaluate the in vivo performance of this novel total meniscal implant. The assessment particularly focused on the implant’s response to long-term physiological loading in a goat model and its chondroprotective capacity in comparison to clinically relevant controls. Study Design: Controlled laboratory study. Methods: Surgery was performed to the stifle joint of 26 female Saanen goats, subdivided into 4 groups: implant, allograft, total meniscectomy, and sham surgery. The sham group’s contralateral joints served as nonoperated controls. After 12 months of follow-up, investigators evaluated implant wear, deformation, and the histopathological condition of the synovium and cartilage. Results: Wear of the implant’s articulating surfaces was minimal, which was confirmed by the absence of wear particles in the synovial fluid. Implant deformation was limited. However, one implant failed by complete tearing of the posterior horn extension. No differences in cartilage histopathological condition were observed for the implant, allograft, and meniscectomy groups. However, locally, the cartilage scores for these groups were significantly worse than those of the nonoperated controls. Conclusion: Whereas this study demonstrated that the novel implant is resistant to wear and that deformation after 12 months of physiological loading is acceptable, reinforcement of the implant horns is necessary to prevent horn failure. Although the implant could not protect the cartilage from developing degenerative changes, the progression of damage was similar in the allograft group. Clinical Relevance: This novel polycarbonate urethane implant may have the potential to become an alternative treatment for symptomatic patients with total meniscectomy.


Isolated genetic deficiencies of individual components of the complement system have been described in man for all the components of the classical pathway and the membrane attack complex as well as for Factor I, Factor H and properdin. It is only for Factor B and Factor D of the alternative pathway that homozygous deficiency states are not so far known. Complement deficiency states provide the most direct way of looking at the role of the complement system in vivo and emphasize the importance of complement in resistance to bacterial infection and in particular to infection with Neisseria . This association is not unexpected since in vitro studies have shown complement to be an efficient enhancer of phagocytosis and inflammation. The particularly frequent occurrence of neisserial infection may be ascribed to the ability of these organisms to survive in phagocytic cells so that the plasma cytolytic activity provided by complement is needed to kill them. On the other hand the strong association between complement deficiencies and immune-complex diseases - especially systemic lupus erythematosus — was unexpected and seems paradoxical in view of the large part played by complement in the pathogenesis of immune complex mediated tissue damage. The paradox can be explained in part by the necessity for an intact complement system in the solubilization and the proper handling of immune complexes. It is also likely that complement deficiency can allow the persistence of low virulence organisms that produce disease solely by an immune complex mechanism. Recently described deficiencies of complement receptors and their effects in vivo are described.


2021 ◽  
Vol 50 (2) ◽  
pp. E6
Author(s):  
Chibawanye I. Ene ◽  
Juan Fueyo ◽  
Frederick F. Lang

Delta-24–based oncolytic viruses are conditional replication adenoviruses developed to selectively infect and replicate in retinoblastoma 1 (Rb)–deficient cancer cells but not normal cell with intact Rb1 pathways. Over the years, there has been a significant evolution in the design of Delta-24 based on a better understanding of the underlying basis for infection, replication, and spread within cancer. One example is the development of Delta-24-RGD (DNX-2401), where the arginine-glycine-aspartate (RGD) domain enhances the infectivity of Delta-24 for cancer cells. DNX-2401 demonstrated objective biological and clinical responses during a phase I window of opportunity clinical trial for recurrent human glioblastoma. In long-term responders (> 3 years), there was evidence of immune infiltration (T cells and macrophages) into the tumor microenvironment with minimal toxicity. Although more in-depth analysis and phase III studies are pending, these results indicate that Delta-24–based adenovirus therapy may induce an antitumor response in glioblastoma, resulting in long-term antitumor immune response. In this review, the authors discuss the preclinical and clinical development of Delta-24 oncolytic adenoviral therapy for glioblastoma and describe structural improvements to Delta-24 that have enhanced its efficacy in vivo. They also highlight ongoing research that attempts to address the remaining obstacles limiting efficacy of Delta-24 adenovirus therapy for glioblastoma.


2007 ◽  
Vol 204 (6) ◽  
pp. 1249-1256 ◽  
Author(s):  
Matthew C. Pickering ◽  
Elena Goicoechea de Jorge ◽  
Rubén Martinez-Barricarte ◽  
Sergio Recalde ◽  
Alfredo Garcia-Layana ◽  
...  

Factor H (FH) is an abundant serum glycoprotein that regulates the alternative pathway of complement-preventing uncontrolled plasma C3 activation and nonspecific damage to host tissues. Age-related macular degeneration (AMD), atypical hemolytic uremic syndrome (aHUS), and membranoproliferative glomerulonephritis type II (MPGN2) are associated with polymorphisms or mutations in the FH gene (Cfh), suggesting the existence of a genotype–phenotype relationship. Although AMD and MPGN2 share pathological similarities with the accumulation of complement-containing debris within the eye and kidney, respectively, aHUS is characterized by renal endothelial injury. This pathological distinction was reflected in our Cfh association analysis, which demonstrated that although AMD and MPGN2 share a Cfh at-risk haplotype, the haplotype for aHUS was unique. FH-deficient mice have uncontrolled plasma C3 activation and spontaneously develop MPGN2 but not aHUS. We show that these mice, transgenically expressing a mouse FH protein functionally equivalent to aHUS-associated human FH mutants, regulate C3 activation in plasma and spontaneously develop aHUS but not MPGN2. These animals represent the first model of aHUS and provide in vivo evidence that effective plasma C3 regulation and the defective control of complement activation on renal endothelium are the critical events in the molecular pathogenesis of FH-associated aHUS.


2020 ◽  
Vol 11 ◽  
Author(s):  
Yuzhou Zhang ◽  
Nicolo Ghiringhelli Borsa ◽  
Dingwu Shao ◽  
Arthur Dopler ◽  
Michael B. Jones ◽  
...  

Factor H (FH), a member of the regulators-of-complement-activation (RCA) family of proteins, circulates in human plasma at concentrations of 180–420 mg/L where it controls the alternative pathway (AP) of complement in the fluid phase and on cell surfaces. When the regulatory function of FH is impaired, complement-mediated tissue injury and inflammation occur, leading to diseases such as atypical hemolytic uremic syndrome (a thrombotic microangiopathy or TMA), C3 glomerulopathy (C3G) and monoclonal gammopathy of renal significance (MGRS). A pathophysiological cause of compromised FH function is the development of autoantibodies to various domains of the FH protein. FH autoantibodies (FHAAs) are identified in 10.9% of patients with aHUS, 3.2% of patients with C3G, and rarely in patients with MGRS. The phenotypic variability of FHAA-mediated disease reflects both the complexity of FH and the epitope specificity of FHAA for select regions of the native protein. In this paper, we have characterized FHAA epitopes in a large cohort of patients diagnosed with TMA, C3G or MGRS. We explore the epitopes recognized by FHAAs in these diseases and the association of FHAAs with the genetic deletion of both copies of the CFHR1 gene to show how these disease phenotypes are associated with this diverse spectrum of autoantibodies.


Blood ◽  
2005 ◽  
Vol 105 (6) ◽  
pp. 2287-2293 ◽  
Author(s):  
Heike Zeitler ◽  
Gudrun Ulrich-Merzenich ◽  
Lothar Hess ◽  
Eligius Konsek ◽  
Christoph Unkrig ◽  
...  

AbstractAcquired hemophilia (AH) is an extremely rare condition in which autoantibodies (inhibitors) against clotting factor VIII induce acute and life-threatening hemorrhagic diathesis because of abnormal blood clotting. The mortality rate of AH is as high as 16%, and current treatment options are associated with adverse side effects. We investigated a therapeutic approach for AH called the modified Bonn-Malmö Protocol (MBMP). The aims of MBMP include suppression of bleeding, permanent elimination of inhibitors, and development of immune tolerance, thereby avoiding long-term reliance on coagulation products. The protocol included immunoadsorption for inhibitor elimination, factor VIII substitution, intravenous immunoglobulin, and immunosuppression. Thirty-five high-titer patients with critical bleeding who underwent MBMP were evaluated. Bleeding was rapidly controlled during 1 or 2 apheresis sessions, and no subsequent bleeding episodes occurred. Inhibitor levels decreased to undetectable levels within a median of 3 days (95% confidence interval [95% CI], 2-4 days), factor substitution was stopped within a median of 12 days (95% CI, 11-17 days), and treatment was completed within a median of 14 days (95% CI, 12-17 days). Long-term follow-up (7 months-7 years) showed an overall response rate of 88% for complete remission (CR). When cancer patients were excluded, the CR rate was 97%.


Cancers ◽  
2021 ◽  
Vol 13 (22) ◽  
pp. 5863
Author(s):  
Hima Patel ◽  
Rosalin Mishra ◽  
Nour Yacoub ◽  
Samar Alanazi ◽  
Mary Kate Kilroy ◽  
...  

The use of BRAF and MEK inhibitors for patients with BRAF-mutant melanoma is limited as patients relapse on treatment as quickly as 6 months due to acquired resistance. We generated trametinib and dabrafenib resistant melanoma (TDR) cell lines to the MEK and BRAF inhibitors, respectively. TDR cells exhibited increased viability and maintenance of downstream p-ERK and p-Akt as compared to parental cells. Receptor tyrosine kinase arrays revealed an increase in p-IGF1R and p-IR in the drug resistant cells versus drug sensitive cells. RNA-sequencing analysis identified IGF1R and INSR upregulated in resistant cell lines compared to parental cells. Analysis of TCGA PanCancer Atlas (skin cutaneous melanoma) showed that patients with a BRAF mutation and high levels of IGF1R and INSR had a worse overall survival. BMS-754807, an IGF1R/IR inhibitor, suppressed cell proliferation along with inhibition of intracellular p-Akt in TDR cells. Dual inhibition of IGF1R and INSR using siRNA reduced cell proliferation. The combination of dabrafenib, trametinib, and BMS-754807 treatment reduced in vivo xenograft tumor growth. Examining the role of IGF1R and IR in mediating resistance to BRAF and MEK inhibitors will expand possible treatment options to aid in long-term success for BRAF-mutant melanoma patients.


2020 ◽  
Author(s):  
Jessica Livingston ◽  
Tina Lee ◽  
Emerson Daniele ◽  
Clara Phillips ◽  
Alexandra Krassikova ◽  
...  

AbstractStroke is the leading cause of adult disability with few treatment options for stroke survivors. Astrocyte reprogramming to neurons enables the targeted in vivo generation of new cells at the site of injury and represents a novel approach for brain repair. A number of studies have demonstrated successful conversion of astrocytes to neurons in various models of brain injury and disease; however, the impact of this strategy on tissue and functional outcome following stroke is not well established. Using AAV delivery of the transcription factor NeuroD1, we reprogrammed astrocytes 7 days after endothelin-1 induced cortical stroke, and studied the long-term cellular and functional outcomes. We found that by 63 days post-stroke, 20% of neurons in the perilesional cortex were reprogrammed. Furthermore, reprogrammed neurons had matured into regionally appropriate neuronal subtypes. Importantly, this treatment was associated with improved functional outcome using the foot fault test and gait analysis. Together, our findings indicate that in vivo reprogramming is a promising regenerative approach for stroke repair.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 637-637 ◽  
Author(s):  
Antonio M. Risitano ◽  
Caterina Pascariello ◽  
Luigi Del Vecchio ◽  
Christopher J Horvath ◽  
Masha Fridkis-Hareli ◽  
...  

Abstract Abstract 637 PNH is characterized by complement (C)-mediated chronic intravascular hemolysis (IVH) due to the absence of CD55 and CD59 on erythrocytes (E) and subsequent impaired C regulation. C alternative pathway (CAP)-derived normal ongoing low level activity (termed the tickover mechanism) is the pivotal initial step, which subsequently leads to C3 fragment (C3frag) deposition on E followed by membrane attack complex (MAC)-mediated IVH. C3frag processing on E proceeds from the initially covalently attached C3b form that is serially converted through proteolysis, mediated on E by factor I and cofactors factor H and complement receptor 1 (CR1), to iC3b and then to C3d as forms that all remain membrane-bound. To develop an improved mechanistic understanding of this process in PNH, which results in C3frag accumulation and extravascular hemolysis (EVH) in vivo during eculizumab (Ecu) treatment, herein we exploited an in vitro model to characterize and dissect C3frag generation after CAP activation on PNH E. We used monoclonal antibodies (mAbs) specific for the different C3 fragments, including C3b/iC3b (mAbs 2C5, 3C11, 3E7, 7C12 and 8E11) and C3b/iC3b/C3d (mAbs A702, 1H8 and 14A10). We also investigated the effect of the C inhibitors (C-Inh) Ecu, 3E7 (a CAP-inhibitory anti-C3b/iC3b mAb) and TT30 (a cell surface-targeted CAP-inhibitory complement receptor 2/factor H fusion protein) on C3frag accumulation and hemolysis of PNH E. E from PNH patients, either naïve or on Ecu treatment, were washed and incubated in ABO-matched acidified normal human serum (aNHS). Assessment of hemolysis and C3 activation and processing was performed by serial flow cytometry analyses of both intact E and E ghosts, as previously described (Risitano et al, Blood 2009; Lindorfer et al, Blood 2010). We first studied E from PNH patients on Ecu, which were known to be C3frag+ using an anti-C3 polyclonal Ab (pAb). Only the anti-C3b/iC3b/C3d mAbs A702, 1H8 and 14A10 demonstrated binding to these PNH E, with a pattern overlapping with that of the anti-C3 pAb, while the anti-C3b/iC3b mAbs 2C5, 3C11, 3E7, 7C12 and 8E11 did not bind. To investigate the kinetics of generation of specific C3frag on PNH E, fresh E from untreated PNH patients that did not show either membrane-bound C3b/iC3b or C3d were exposed in vitro to aNHSs. Analysis at 1h and 24h revealed that intact E, which remained C3frag-negative, progressively decreased and finally disappeared, being transformed into C3b/iC3b+ and C3d+ ghosts. In the presence of Ecu (using serum from patients on Ecu treatment, drawn within 1h of dosing), the same experiments revealed a dramatic reduction of hemolysis, but residual CAP-mediated hemolysis in a process of pharmacodynamic breakthrough was confirmed by the presence of C3b/iC3b+, C3d+, CD59− E ghosts. More interestingly, intact E showed substantial C3frag deposition which progressed over time, initially characterized by the presence of C3b/iC3b and C3d, and subsequently converting after 24h exclusively into C3d+ E, with an identical phenotype as found in C3frag+ PNH E obtained from patients on Ecu. This result demonstrates that PNH E, despite exhibiting only C3d when recovered from patients, pass through an earlier phase where C3b/iC3b frag are present that can interact with their cognate receptors on fixed cells in liver and spleen during EVH in vivo. Both 3E7 and TT30 completely inhibited hemolysis, with an IC50 of 120 μg/mL for 3E7 and 30 μg/mL for TT30; consistent with their mechanism of action, both 3E7 and TT30 bound to PNH RBCs. No C3b/iC3b nor C3d was detected on surviving PNH E at any time, indicating that 3E7 and TT30 effectively inhibit the earliest phases of CAP activation involved in EVH. Similar results were obtained when E from PNH patients on Ecu were challenged in vitro with aNHS in the presence or absence of C-Inh. In conclusion, we demonstrate that CAP-mediated C3b/iC3b fixation to PNH RBCs is the early event leading to IVH or, in presence of Ecu, to C3frag deposition and subsequent EVH in vivo. Unlike Ecu, use of 3E7 or TT30 resulted in complete inhibition of hemolysis as well as CAP-mediated C3 activation, preventing C3b/iC3b and C3d deposition on intact PNH E. Thus, use of this model will allow assessment of the roles of both endogenous and therapeutic C regulators in CAP activation and C3 processing (and subsequent EVH) in PNH. Preclinical data suggest that TT30 is an optimal candidate agent to assess in vivo the effect of CAP inhibition in PNH patients. Disclosures: Risitano: Taligen Therapeutics: Consultancy, Research Funding. Horvath:Taligen Therapeutics: Employment. Fridkis-Hareli:Taligen Therapeutics: Employment. Holers:Taligen Therapeutics: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties.


Sign in / Sign up

Export Citation Format

Share Document