Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Pharmacol Exp Ther ; 351(3): 527-37, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25232192

ABSTRACT

Anti-factor D (AFD; FCFD4514S, lampalizumab) is a humanized IgG Fab fragment directed against factor D (fD), a rate-limiting serine protease in the alternative complement pathway (AP). Evaluation of AFD as a potential intravitreal (IVT) therapeutic for dry age-related macular degeneration patients with geographic atrophy (GA) is ongoing. However, it is unclear whether IVT administration of AFD can affect systemic AP activation and potentially compromise host-immune responses. We characterized the pharmacologic properties of AFD and assessed the effects of AFD administered IVT (2 or 20 mg) or intravenous (0.2, 2, or 20 mg) on systemic complement activity in cynomolgus monkeys. For the IVT groups, serum AP activity was reduced for the 20 mg dose group between 2 and 6 hours postinjection. For the intravenous groups, AFD inhibited systemic AP activity for periods of time ranging from 5 minutes (0.2 mg group) to 3 hours (20 mg group). Interestingly, the concentrations of total serum fD increased up to 10-fold relative to predose levels following administration of AFD. Furthermore, AFD was found to inhibit systemic AP activity only when the molar concentration of AFD exceeded that of fD. This occurred in cynomolgus monkeys at serum AFD levels ≥2 µg/ml, a concentration 8-fold greater than the maximum serum concentration observed following a single 10 mg IVT dose in a clinical investigation in patients with GA. Based on these findings, the low levels of serum AFD resulting from IVT administration of a clinically relevant dose are not expected to appreciably affect systemic AP activity.


Subject(s)
Complement C3a/antagonists & inhibitors , Complement Factor D/antagonists & inhibitors , Immunoglobulin Fab Fragments/administration & dosage , Macular Degeneration/drug therapy , Animals , Cattle , Complement C3a/immunology , Complement Factor D/immunology , Dose-Response Relationship, Drug , Female , Humans , Immunoglobulin Fab Fragments/immunology , Intravitreal Injections , Macaca fascicularis , Macular Degeneration/blood , Macular Degeneration/immunology , Male , Mice , Treatment Outcome
2.
Invest Ophthalmol Vis Sci ; 53(10): 6628-37, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-22930722

ABSTRACT

PURPOSE: To determine if the progression of age-related macular degeneration (AMD) is associated with complement activation in the eye. METHODS: Immunohistochemistry and ELISAs were used to determine the distribution, concentration, and activation of the alternative pathway complement proteases factor B (FB) and factor D (FD) and the central complement protein C3 in genotyped human postmortem donor eyes graded as having no or minimal drusen (category 1; controls), large drusen (category 3), and large drusen with advanced AMD (category 4). RESULTS: C3, FB, and FD were present in vitreous and Bruch's membrane choroid (BM/C) interface of the macula of eyes in all tested AMD severity categories (n = 100). C3, FB, and FD were predominantly located to the choroidal vasculature and Bruch's membrane and, together with the serum proteins transferrin and albumin, elevated in BM/C extracts of category 4 eyes (n = 23) compared with category 1 eyes (n = 24). A significant increase in FB activation was found only in vitreous of category 4 eyes (n = 23) compared with category 1 eyes (n = 25). Genetic variants of complement factor H (CFH), C3, C2, and FB associated with increased risk of AMD were correlated with alternative pathway complement activation in vitreous, but not with complement proteins in BM/C protein extracts. CONCLUSIONS: Increased activation of the alternative complement pathway in vitreous was controlled by disease stage and genetic variation in the complement pathway, supporting a role for complement activation in AMD disease pathogenesis.


Subject(s)
Complement C3/metabolism , Complement Factor B/metabolism , Complement Factor D/metabolism , Complement Pathway, Alternative/genetics , Macular Degeneration/genetics , Vitreous Body/metabolism , Aged , Aged, 80 and over , Bruch Membrane/metabolism , Choroid/metabolism , Complement Activation , Complement C3/genetics , Complement Factor B/genetics , Complement Factor D/genetics , Complement Factor H/genetics , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Genotype , Humans , Immunohistochemistry , Macular Degeneration/metabolism , Male , Middle Aged , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Tissue Donors
3.
J Biol Chem ; 284(51): 35605-11, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-19833734

ABSTRACT

CRIg is a recently discovered complement C3 receptor expressed on a subpopulation of tissue-resident macrophages. The extracellular IgV domain of CRIg (CRIg-ECD) holds considerable promise as a potential therapeutic because it selectively inhibits the alternative pathway of complement by binding to C3b and inhibiting proteolytic activation of C3 and C5. However, CRIg binds weakly to the convertase subunit C3b (K(D) = 1.1 microm), and thus a relatively high concentration of protein is required to reach nearly complete complement inhibition. To improve therapeutic efficacy while minimizing risk of immunogenicity, we devised a phage display strategy to evolve a high affinity CRIg-ECD variant with a minimal number of mutations. Using the crystal structure of CRIg in complex with C3b as a guide for library design, we isolated a CRIg-ECD double mutant (Q64R/M86Y, CRIg-v27) that showed increased binding affinity and improved complement inhibitory activity relative to CRIg-ECD. In a mouse model of arthritis, treatment with a Fc fusion of CRIg-v27 resulted in a significant reduction in clinical scores compared with treatment with an Fc fusion of CRIg-ECD. This study clearly illustrates how phage display technology and structural information can be combined to generate proteins with nearly natural sequences that act as potent complement inhibitors with greatly improved therapeutic efficacy.


Subject(s)
Arthritis/drug therapy , Receptors, Complement 3b/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Amino Acid Substitution , Animals , Arthritis/metabolism , Complement C3b/genetics , Complement C3b/metabolism , Complement C5/genetics , Complement C5/metabolism , Complement Pathway, Alternative/drug effects , Disease Models, Animal , Humans , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mutation, Missense , Protein Structure, Tertiary/physiology , Rabbits , Receptors, Complement 3b/chemistry , Receptors, Complement 3b/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Structure-Activity Relationship
4.
J Biol Chem ; 284(16): 10473-9, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19196712

ABSTRACT

Amplification of the complement cascade through the alternative pathway can lead to excessive inflammation. Targeting C3b, a component central to the alternative pathway of complement, provides a powerful approach to inhibit complement-mediated immune responses and tissue injury. In the present study, phage display technology was employed to generate an antibody that selectively recognizes C3b but not the non-activated molecule C3. The crystal structure of C3b in complex with a Fab fragment of this antibody (S77) illustrates the structural basis for this selectivity. Cleavage of C3 to C3b results in a plethora of structural changes within C3, including the rearrangement of macroglobulin domain 6 enabling binding of S77 to the adjacent macroglobulin domain 7 domain. S77 blocks binding of factor B to C3b inhibiting the first step in the formation of the alternative pathway C3 convertase. In addition, S77 inhibits C5 binding to C3b. This results in significantly reduced formations of anaphylatoxins and membrane-attack complexes. This study for the first time demonstrates the structural basis for complement inhibition by a C3b-selective antibody and provides insights into the molecular mechanisms of alternative pathway complement activation.


Subject(s)
Antibodies , Complement C3b/immunology , Complement Pathway, Alternative/physiology , Immunoglobulin Fab Fragments , Protein Conformation , Animals , Antibodies/chemistry , Antibodies/metabolism , Complement C3 Convertase, Alternative Pathway/metabolism , Complement C3b/chemistry , Complement C3b/genetics , Complement C3b/metabolism , Complement C5 Convertase, Alternative Pathway/metabolism , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Stability , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Macaca mulatta , Models, Molecular , Molecular Sequence Data , Peptide Library , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Receptors, Complement 3b/chemistry , Receptors, Complement 3b/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...