Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 34
Filtrer
1.
Front Immunol ; 12: 712572, 2021.
Article de Anglais | MEDLINE | ID: mdl-34566967

RÉSUMÉ

The complement system is central to first-line defense against invading pathogens. However, excessive complement activation and/or the loss of complement regulation contributes to the development of autoimmune diseases, systemic inflammation, and thrombosis. One of the three pathways of the complement system, the alternative complement pathway, plays a vital role in amplifying complement activation and pathway signaling. Complement factor D, a serine protease of this pathway that is required for the formation of C3 convertase, is the rate-limiting enzyme. In this review, we discuss the function of factor D within the alternative pathway and its implication in both healthy physiology and disease. Because the alternative pathway has a role in many diseases that are characterized by excessive or poorly mediated complement activation, this pathway is an enticing target for effective therapeutic intervention. Nonetheless, although the underlying disease mechanisms of many of these complement-driven diseases are quite well understood, some of the diseases have limited treatment options or no approved treatments at all. Therefore, in this review we explore factor D as a strategic target for advancing therapeutic control of pathological complement activation.


Sujet(s)
Facteur D du complément/antagonistes et inhibiteurs , Voie alterne d'activation du complément/effets des médicaments et des substances chimiques , Thérapie moléculaire ciblée , Tissu adipeux/métabolisme , Vieillissement/immunologie , Vascularites associées aux anticorps anti-cytoplasme des neutrophiles/traitement médicamenteux , Vascularites associées aux anticorps anti-cytoplasme des neutrophiles/immunologie , Anticorps monoclonaux humanisés/usage thérapeutique , Maladies auto-immunes/immunologie , Maladies auto-immunes/thérapie , Facteur D du complément/biosynthèse , Facteur D du complément/déficit , Facteur D du complément/physiologie , Métabolisme énergétique , Atrophie géographique/génétique , Atrophie géographique/immunologie , Hémoglobinurie paroxystique/traitement médicamenteux , Hémoglobinurie paroxystique/génétique , Hémoglobinurie paroxystique/immunologie , Hépatocytes , Humains , Maladies du rein/immunologie , Foie/traumatismes , Oligonucléotides antisens/usage thérapeutique , Peptides cycliques/usage thérapeutique , Phagocytose
2.
Front Immunol ; 12: 690821, 2021.
Article de Anglais | MEDLINE | ID: mdl-34177949

RÉSUMÉ

Complement factor B (FB) mutant variants are associated with excessive complement activation in kidney diseases such as atypical hemolytic uremic syndrome (aHUS), C3 glomerulopathy and membranoproliferative glomerulonephritis (MPGN). Patients with aHUS are currently treated with eculizumab while there is no specific treatment for other complement-mediated renal diseases. In this study the phenotype of three FB missense variants, detected in patients with aHUS (D371G and E601K) and MPGN (I242L), was investigated. Patient sera with the D371G and I242L mutations induced hemolysis of sheep erythrocytes. Mutagenesis was performed to study the effect of factor D (FD) inhibition on C3 convertase-induced FB cleavage, complement-mediated hemolysis, and the release of soluble C5b-9 from glomerular endothelial cells. The FD inhibitor danicopan abrogated C3 convertase-associated FB cleavage to the Bb fragment in patient serum, and of the FB constructs, D371G, E601K, I242L, the gain-of-function mutation D279G, and the wild-type construct, in FB-depleted serum. Furthermore, the FD-inhibitor blocked hemolysis induced by the D371G and D279G gain-of-function mutants. In FB-depleted serum the D371G and D279G mutants induced release of C5b-9 from glomerular endothelial cells that was reduced by the FD-inhibitor. These results suggest that FD inhibition can effectively block complement overactivation induced by FB gain-of-function mutations.


Sujet(s)
Syndrome hémolytique et urémique atypique/immunologie , Activation du complément , Facteur B du complément/génétique , Facteur D du complément/antagonistes et inhibiteurs , Glomérulonéphrite membranoproliférative/immunologie , Animaux , Syndrome hémolytique et urémique atypique/génétique , Enfant , Complement C3-C5 Convertases/immunologie , Complément C3b/immunologie , Facteur B du complément/immunologie , Facteur D du complément/immunologie , Cellules endothéliales/immunologie , Érythrocytes , Femelle , Glomérulonéphrite membranoproliférative/génétique , Hémolyse , Humains , Nourrisson , Glomérule rénal/cytologie , Mâle , Adulte d'âge moyen , Mutation , Phénotype , Lapins , Ovis
4.
Blood ; 136(18): 2080-2089, 2020 10 29.
Article de Anglais | MEDLINE | ID: mdl-32877502

RÉSUMÉ

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a highly contagious respiratory virus that can lead to venous/arterial thrombosis, stroke, renal failure, myocardial infarction, thrombocytopenia, and other end-organ damage. Animal models demonstrating end-organ protection in C3-deficient mice and evidence of complement activation in humans have led to the hypothesis that SARS-CoV-2 triggers complement-mediated endothelial damage, but the mechanism is unclear. Here, we demonstrate that the SARS-CoV-2 spike protein (subunit 1 and 2), but not the N protein, directly activates the alternative pathway of complement (APC). Complement-dependent killing using the modified Ham test is blocked by either C5 or factor D inhibition. C3 fragments and C5b-9 are deposited on TF1PIGAnull target cells, and complement factor Bb is increased in the supernatant from spike protein-treated cells. C5 inhibition prevents the accumulation of C5b-9 on cells, but not C3c; however, factor D inhibition prevents both C3c and C5b-9 accumulation. Addition of factor H mitigates the complement attack. In conclusion, SARS-CoV-2 spike proteins convert nonactivator surfaces to activator surfaces by preventing the inactivation of the cell-surface APC convertase. APC activation may explain many of the clinical manifestations (microangiopathy, thrombocytopenia, renal injury, and thrombophilia) of COVID-19 that are also observed in other complement-driven diseases such as atypical hemolytic uremic syndrome and catastrophic antiphospholipid antibody syndrome. C5 inhibition prevents accumulation of C5b-9 in vitro but does not prevent upstream complement activation in response to SARS-CoV-2 spike proteins.


Sujet(s)
Betacoronavirus , Facteur D du complément/antagonistes et inhibiteurs , Inhibiteurs du complément/pharmacologie , Voie alterne d'activation du complément/effets des médicaments et des substances chimiques , Glycoprotéine de spicule des coronavirus/pharmacologie , Lignée cellulaire , Activation du complément/effets des médicaments et des substances chimiques , Complément C3/métabolisme , Complément C5/antagonistes et inhibiteurs , Facteur H du complément/métabolisme , Complexe d'attaque membranaire du complément/métabolisme , Humains , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus/physiologie
5.
Front Immunol ; 10: 1007, 2019.
Article de Anglais | MEDLINE | ID: mdl-31156618

RÉSUMÉ

Ocular inflammation is a defining feature of sight threating diseases and its dysregulation can catalyze and or propagate ocular neurodegenerative maladies such as age-related macular degeneration (AMD). The complement system, an intrinsic component of the innate immunity, has an integral role in maintaining immune-surveillance and homeostasis in the ocular microenvironment; however, overstimulation can drive ocular inflammatory diseases. The mechanism for complement disease propagation in AMD is not fully understood, although there is accumulating evidence showing that targeted modulation of complement-specific proteins has the potential to become a viable therapeutic approach. To date, a major focus of complement therapeutics has been on targeting the alternative complement system in AMD. Recent studies have outlined potential complement cascade inhibitors that might mitigate AMD disease progression. First-in-class complement inhibitors target the modulation of complement proteins C3, C5, factor B, factor D, and properdin. Herein, we will summarize ocular inflammation in the context of AMD disease progression, current clinical outcomes and complications of complement-mediated therapeutics. Given the need for additional therapeutic approaches for ocular inflammatory diseases, targeted complement modulation has emerged as a leading candidate for eliminating inflammation-driven ocular maladies.


Sujet(s)
Antigènes CD59/antagonistes et inhibiteurs , Complément C3/antagonistes et inhibiteurs , Complément C5/antagonistes et inhibiteurs , Facteur D du complément/antagonistes et inhibiteurs , Dégénérescence maculaire/traitement médicamenteux , Thérapie moléculaire ciblée/méthodes , Properdine/antagonistes et inhibiteurs , Animaux , Antigènes CD59/métabolisme , Activation du complément/effets des médicaments et des substances chimiques , Activation du complément/immunologie , Complément C3/métabolisme , Complément C5/métabolisme , Facteur D du complément/métabolisme , Humains , Immunité innée , Inflammation/traitement médicamenteux , Inflammation/immunologie , Dégénérescence maculaire/immunologie , Dégénérescence maculaire/métabolisme , Properdine/métabolisme
6.
J Med Chem ; 62(9): 4656-4668, 2019 05 09.
Article de Anglais | MEDLINE | ID: mdl-30995036

RÉSUMÉ

Complement factor D (FD), a highly specific S1 serine protease, plays a central role in the amplification of the alternative complement pathway (AP) of the innate immune system. Dysregulation of AP activity predisposes individuals to diverse disorders such as age-related macular degeneration, atypical hemolytic uremic syndrome, membranoproliferative glomerulonephritis type II, and paroxysmal nocturnal hemoglobinuria. Previously, we have reported the screening efforts and identification of reversible benzylamine-based FD inhibitors (1 and 2) binding to the open active conformation of FD. In continuation of our drug discovery program, we designed compounds applying structure-based approaches to improve interactions with FD and gain selectivity against S1 serine proteases. We report herein the design, synthesis, and medicinal chemistry optimization of the benzylamine series culminating in the discovery of 12, an orally bioavailable and selective FD inhibitor. 12 demonstrated systemic suppression of AP activation in a lipopolysaccharide-induced AP activation model as well as local ocular suppression in intravitreal injection-induced AP activation model in mice expressing human FD.


Sujet(s)
Benzylamines/pharmacologie , Voie alterne d'activation du complément/effets des médicaments et des substances chimiques , Inhibiteurs de la sérine protéinase/pharmacologie , Animaux , Benzylamines/synthèse chimique , Benzylamines/métabolisme , Sites de fixation , Facteur D du complément/antagonistes et inhibiteurs , Facteur D du complément/composition chimique , Facteur D du complément/métabolisme , Chiens , Conception de médicament , Humains , Souris de lignée C57BL , Souris transgéniques , Simulation de docking moléculaire , Conformation des protéines , Rats , Inhibiteurs de la sérine protéinase/synthèse chimique , Inhibiteurs de la sérine protéinase/métabolisme
7.
Biofactors ; 45(4): 556-562, 2019 Jul.
Article de Anglais | MEDLINE | ID: mdl-31026383

RÉSUMÉ

Aging is associated with a decrease of extracellular matrix and an increase of senescent cells in the dermal layer. Here, to examine whether and how senescent cells are involved in aging-related deterioration of the dermal layer, we cocultured dermal young fibroblasts (low-passage number) with senescent cells (high-passage number) in Transwells, in which the two cell types are separated by a semipermeable membrane. Young fibroblasts in coculture showed decreased collagen type I alpha 1 chain and elastin gene expression, and increased matrix metalloproteinase 1 (MMP1) gene expression. To identify causative factors, we compared gene expression of young and senescent cells and selected candidate secretory factors whose expression was increased by ≥2.5 in senescent fibroblasts. Then, we used siRNAs to knock down each of the 11 candidate genes in senescent fibroblasts in the coculture system. Knockdown of complement factor D (CFD) in senescent fibroblasts significantly reduced the increase of MMP1 in the cocultured young fibroblasts. In monocultures, treatment of young fibroblasts with CFD resulted in increased MMP1 gene expression, while knockdown of CFD in senescent fibroblasts decreased MMP1 gene expression. In addition, production of CFD was increased in culture medium of untreated senescent fibroblasts. Furthermore, CFD gene and protein expression were increased in the dermal layer of skin specimens from aged subjects (>70 years old), compared to young subjects (<20 years old). Overall, these results suggest that senescent cells negatively influence matrix production and promote degradation of nearby fibroblasts in the dermal layer, in part through secretion of CFD.


Sujet(s)
Matrice extracellulaire/métabolisme , Fibroblastes/métabolisme , Matrix metalloproteinase 1/génétique , Prolifération cellulaire , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Techniques de coculture , Collagène de type I/génétique , Collagène de type I/métabolisme , Chaine alpha-1 du collagène de type I , Facteur D du complément/antagonistes et inhibiteurs , Facteur D du complément/génétique , Facteur D du complément/métabolisme , Facteur D du complément/pharmacologie , Chambres de culture à diffusion , Élastine/génétique , Élastine/métabolisme , Matrice extracellulaire/composition chimique , Matrice extracellulaire/effets des médicaments et des substances chimiques , Fibroblastes/cytologie , Fibroblastes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes , Humains , Matrix metalloproteinase 1/métabolisme , Culture de cellules primaires , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Transduction du signal
8.
Mol Pharm ; 16(1): 86-95, 2019 01 07.
Article de Anglais | MEDLINE | ID: mdl-30444371

RÉSUMÉ

The collection of aqueous humor (phase 1 b/2 Mahalo study) from patients dosed intravitreally with anti-factor D (AFD; FCFD4514S, lampalizumab), a humanized antibody fragment previously under investigation to treat geographic atrophy (GA) secondary to age-related macular degeneration, presented a unique opportunity to examine AFD properties in clinical samples. We investigated AFD stability and target-binding characteristics to set up strategies for engineering and evaluating optimized molecules that enable less frequent dosing. Two variants, AFD.v8 and AFD.v14, were evaluated as alternatives to AFD for longer-acting treatments. Mass spectrometry, surface plasmon resonance, and immunoassay were used to assess AFD stability and binding activity in aqueous humor samples from Mahalo patients. In vitro stability and binding activity of AFD, AFD.v8, and AFD.v14 were assessed in human vitreous humor versus buffer at 37 °C over 16 weeks and in vivo in rabbits over 28 days along with pharmacokinetic determinations. In human aqueous humor, AFD specific binding was >85% through 30 days, and deamidation was <3% through 60 days, consistent with the AFD stability and binding activity in vitreous humor from humans in vitro and rabbits in vivo. Target binding, stability, and rabbit pharmacokinetic parameters of AFD.v8 and AFD.v14 were similar to those of AFD. Physiological stability and activity of AFD translated across in vitro and in vivo studies in humans and rabbits. The two variants AFD.v8 and AFD.v14 demonstrated comparable potency and pharmacokinetics. These findings, along with previously demonstrated improved solubility of AFD.v8 and AFD.v14, provide proof-of-concept for developing other similar long-acting therapeutic variants.


Sujet(s)
Humeur aqueuse/métabolisme , Facteur D du complément/antagonistes et inhibiteurs , Fragments Fab d'immunoglobuline/métabolisme , Animaux , Atrophie géographique/métabolisme , Humains , Dosage immunologique , Fragments Fab d'immunoglobuline/usage thérapeutique , Dégénérescence maculaire/métabolisme , Mâle , Spectrométrie de masse , Lapins , Résonance plasmonique de surface , Corps vitré/métabolisme
9.
JAMA Ophthalmol ; 136(6): 666-677, 2018 06 01.
Article de Anglais | MEDLINE | ID: mdl-29801123

RÉSUMÉ

Importance: Geographic atrophy (GA) secondary to age-related macular degeneration is a leading cause of visual disability in older individuals. A phase 2 trial suggested that lampalizumab, a selective complement factor D inhibitor, reduced the rate of GA enlargement, warranting phase 3 trials. Objective: To assess the safety and efficacy of lampalizumab vs sham procedure on enlargement of GA. Design, Setting, and Participants: Two identically designed phase 3 double-masked, randomized, sham-controlled clinical trials, Chroma and Spectri, enrolled participants from August 28, 2014, to October 6, 2016, at 275 sites in 23 countries. Participants were aged 50 years or older, with bilateral GA and no prior or active choroidal neovascularization in either eye and GA lesions in the study eye measuring 2.54 to 17.78 mm2 with diffuse or banded fundus autofluorescence patterns. Interventions: Participants were randomized 2:1:2:1 to receive 10 mg of intravitreous lampalizumab every 4 weeks, sham procedure every 4 weeks, 10 mg of lampalizumab every 6 weeks, or sham procedure every 6 weeks, through 96 weeks. Main Outcomes and Measures: Safety and efficacy assessed as mean change from baseline in GA lesion area at week 48 from centrally read fundus autofluorescence images of the lampalizumab arms vs pooled sham arms, in the intent-to-treat population and by complement factor I-profile genetic biomarker. Results: A total of 906 participants (553 women and 353 men; mean [SD] age, 78.1 [8.1] years) were enrolled in Chroma and 975 participants (578 women and 397 men; mean [SD] age, 77.9 [8.1] years) were enrolled in Spectri; 1733 of the 1881 participants (92.1%) completed the studies through 48 weeks. The adjusted mean increases in GA lesion area from baseline at week 48 were 1.93 to 2.09 mm2 across all groups in both studies. Differences in adjusted mean change in GA lesion area (lampalizumab minus sham) were -0.02 mm2 (95% CI, -0.21 to 0.16 mm2; P = .80) for lampalizumab every 4 weeks in Chroma, 0.16 mm2 (95% CI, 0.00-0.31 mm2; P = .048) for lampalizumab every 4 weeks in Spectri, 0.05 mm2 (95% CI, -0.13 to 0.24 mm2; P = .59) for lampalizumab every 6 weeks in Chroma, and 0.09 mm2 (95% CI, -0.07 to 0.24 mm2; P = .27) for lampalizumab every 6 weeks in Spectri. No benefit of lampalizumab was observed across prespecified subgroups, including by complement factor I-profile biomarker. Endophthalmitis occurred after 5 of 12 447 injections (0.04%) or in 5 of 1252 treated participants (0.4%) through week 48. Conclusions and Relevance: In Chroma and Spectri, the largest studies of GA conducted to date, lampalizumab did not reduce GA enlargement vs sham during 48 weeks of treatment. Results highlight the substantial and consistent enlargement of GA, at a mean of approximately 2 mm2 per year. Trial Registration: ClinicalTrials.gov Identifier: NCT02247479 and NCT02247531.


Sujet(s)
Atrophie géographique/traitement médicamenteux , Fragments Fab d'immunoglobuline/usage thérapeutique , Dégénérescence maculaire/complications , Sujet âgé , Sujet âgé de 80 ans ou plus , Facteur D du complément/antagonistes et inhibiteurs , Méthode en double aveugle , Femelle , Angiographie fluorescéinique , Atrophie géographique/diagnostic , Atrophie géographique/étiologie , Humains , Fragments Fab d'immunoglobuline/effets indésirables , Injections intravitréennes , Mâle , Adulte d'âge moyen , Études prospectives , Résultat thérapeutique , Acuité visuelle
10.
Invest Ophthalmol Vis Sci ; 59(2): 940-951, 2018 02 01.
Article de Anglais | MEDLINE | ID: mdl-29450541

RÉSUMÉ

Purpose: Genome-wide association studies suggest a role for the complement system in age-related macular degeneration (AMD). We characterized ocular complement activation and evaluated a complement factor D (FD) neutralizing antibody. Methods: Mice were treated with toll-like receptor (TLR) ligands, intravitreal injection (IVT), or corneal debridement. Levels of complement proteins and mRNA were measured. A FD neutralizing antibody was administered IVT into eyes of rabbits that were challenged with LPS (lipopolysaccharide) administered intravenously. Results: Levels of C3 and factor B (FB) mRNA and protein in the eye were increased following intraperitoneal injection of TLR4 ligand LPS. Increased levels of C3 and FB breakdown products were observed in both eye tissues and plasma. Complement activation products were markedly reduced in C3-/- and Cfb-/- mice challenged with LPS. Ocular complement levels were also elevated in mice treated systemically with TLR2 and -3 ligands, injured by IVT injection or corneal debridement, or even in normal aging. IVT administration of a complement FD neutralizing antibody in rabbits inhibited LPS-induced complement activation in the posterior segment of the eye, but not in the anterior segment of the eye or in plasma. Conclusions: Systemic TLR stimulation and eye tissue injury induced time-dependent alternative complement pathway activation in the eye. Ocular complement levels were also gradually elevated during aging. An anti-FD antibody IVT potently inhibited LPS-induced complement activation in the posterior segment of the eye. This study provides insights into the dynamic profile of ocular complement activation, which is valuable for complement research in eye diseases and for developing complement therapeutics for AMD.


Sujet(s)
Anticorps neutralisants/pharmacologie , Facteur D du complément/antagonistes et inhibiteurs , Voie alterne d'activation du complément/physiologie , Inflammation/immunologie , Modèles animaux , Animaux , Technique de Western , Complément C3/métabolisme , Facteur B du complément/métabolisme , Femelle , Injections péritoneales , Injections intravitréennes , Lipopolysaccharides/pharmacologie , Souris , Souris de lignée C57BL , ARN messager/génétique , Lapins , Récepteur de type Toll-4/métabolisme
11.
J Med Chem ; 60(13): 5717-5735, 2017 07 13.
Article de Anglais | MEDLINE | ID: mdl-28621538

RÉSUMÉ

The highly specific S1 serine protease factor D (FD) plays a central role in the amplification of the complement alternative pathway (AP) of the innate immune system. Genetic associations in humans have implicated AP activation in age-related macular degeneration (AMD), and AP dysfunction predisposes individuals to disorders such as paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (aHUS). The combination of structure-based hit identification and subsequent optimization of the center (S)-proline-based lead 7 has led to the discovery of noncovalent reversible and selective human factor D (FD) inhibitors with drug-like properties. The orally bioavailable compound 2 exerted excellent potency in 50% human whole blood in vitro and blocked AP activity ex vivo after oral administration to monkeys as demonstrated by inhibition of membrane attack complex (MAC) formation. Inhibitor 2 demonstrated sustained oral and ocular efficacy in a model of lipopolysaccharide (LPS)-induced systemic AP activation in mice expressing human FD.


Sujet(s)
Facteur D du complément/antagonistes et inhibiteurs , Voie alterne d'activation du complément/effets des médicaments et des substances chimiques , Proline/analogues et dérivés , Proline/pharmacologie , Administration par voie orale , Animaux , Syndrome hémolytique et urémique atypique/traitement médicamenteux , Syndrome hémolytique et urémique atypique/immunologie , Facteur D du complément/immunologie , Complexe d'attaque membranaire du complément/antagonistes et inhibiteurs , Complexe d'attaque membranaire du complément/immunologie , Femelle , Haplorhini , Humains , Macaca fascicularis , Dégénérescence maculaire/traitement médicamenteux , Dégénérescence maculaire/immunologie , Mâle , Souris , Proline/administration et posologie , Proline/pharmacocinétique
12.
Sci Transl Med ; 9(395)2017 06 21.
Article de Anglais | MEDLINE | ID: mdl-28637922

RÉSUMÉ

Geographic atrophy is an advanced form of age-related macular degeneration (AMD) and a leading cause of vision loss for which there are no approved treatments. Genetic studies in AMD patients have implicated dysregulation of the alternative complement pathway in the pathogenesis of geographic atrophy. Lampalizumab is a potential therapeutic that targets complement factor D, a pivotal activator of the alternative complement pathway. The MAHALO phase 2 clinical trial was a multicenter, randomized, controlled study that evaluated lampalizumab administered by intravitreal injection monthly (n = 42) and every other month (n = 41) versus sham control (n = 40) in patients with geographic atrophy secondary to AMD. The primary endpoint was the mean change in lesion area from baseline to month 18 as measured by fundus autofluorescence. Specific AMD-associated genetic polymorphisms were also analyzed. The MAHALO study met its primary efficacy endpoint with an acceptable safety profile; monthly lampalizumab treatment demonstrated a 20% reduction in lesion area progression versus sham control [80% confidence interval (CI), 4 to 37%]. A more substantial monthly treatment benefit of 44% reduction in geographic atrophy area progression versus sham control (95% CI, 15 to 73%) was observed in a subgroup of complement factor I (CFI) risk-allele carriers (57% of the patients analyzed were CFI risk-allele carriers). The MAHALO study shows a potential treatment effect in patients with geographic atrophy and supports therapeutic targeting of the alternative complement pathway for treating AMD pathogenesis.


Sujet(s)
Atrophie géographique/traitement médicamenteux , Atrophie géographique/métabolisme , Fragments Fab d'immunoglobuline/usage thérapeutique , Dégénérescence maculaire/traitement médicamenteux , Dégénérescence maculaire/métabolisme , Sujet âgé , Facteur D du complément/antagonistes et inhibiteurs , Facteur D du complément/métabolisme , Voie alterne d'activation du complément , Évolution de la maladie , Femelle , Atrophie géographique/anatomopathologie , Humains , Dégénérescence maculaire/anatomopathologie , Mâle , Adulte d'âge moyen
13.
Haematologica ; 102(3): 466-475, 2017 03.
Article de Anglais | MEDLINE | ID: mdl-27810992

RÉSUMÉ

Paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome are diseases of excess activation of the alternative pathway of complement that are treated with eculizumab, a humanized monoclonal antibody against the terminal complement component C5. Eculizumab must be administered intravenously, and moreover some patients with paroxysmal nocturnal hemoglobinuria on eculizumab have symptomatic extravascular hemolysis, indicating an unmet need for additional therapeutic approaches. We report the activity of two novel small-molecule inhibitors of the alternative pathway component Factor D using in vitro correlates of both paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome. Both compounds bind human Factor D with high affinity and effectively inhibit its proteolytic activity against purified Factor B in complex with C3b. When tested using the traditional Ham test with cells from paroxysmal nocturnal hemoglobinuria patients, the Factor D inhibitors significantly reduced complement-mediated hemolysis at concentrations as low as 0.01 µM. Additionally the compound ACH-4471 significantly decreased C3 fragment deposition on paroxysmal nocturnal hemoglobinuria erythrocytes, indicating a reduced potential relative to eculizumab for extravascular hemolysis. Using the recently described modified Ham test with serum from patients with atypical hemolytic uremic syndrome, the compounds reduced the alternative pathway-mediated killing of PIGA-null reagent cells, thus establishing their potential utility for this disease of alternative pathway of complement dysregulation and validating the modified Ham test as a system for pre-clinical drug development for atypical hemolytic uremic syndrome. Finally, ACH-4471 blocked alternative pathway activity when administered orally to cynomolgus monkeys. In conclusion, the small-molecule Factor D inhibitors show potential as oral therapeutics for human diseases driven by the alternative pathway of complement, including paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome.


Sujet(s)
Syndrome hémolytique et urémique atypique/étiologie , Syndrome hémolytique et urémique atypique/métabolisme , Facteur D du complément/antagonistes et inhibiteurs , Inhibiteurs du complément/pharmacologie , Voie alterne d'activation du complément/effets des médicaments et des substances chimiques , Voie alterne d'activation du complément/immunologie , Hémoglobinurie paroxystique/étiologie , Hémoglobinurie paroxystique/métabolisme , Adulte , Sujet âgé , Animaux , Syndrome hémolytique et urémique atypique/diagnostic , Syndrome hémolytique et urémique atypique/traitement médicamenteux , Marqueurs biologiques , Complément C3/immunologie , Complément C3/métabolisme , Facteur D du complément/immunologie , Facteur D du complément/métabolisme , Inhibiteurs du complément/administration et posologie , Cytotoxicité immunologique , Modèles animaux de maladie humaine , Érythrocytes/immunologie , Érythrocytes/métabolisme , Femelle , Hémoglobinurie paroxystique/diagnostic , Hémoglobinurie paroxystique/traitement médicamenteux , Hémolyse , Humains , Macaca fascicularis , Mâle , Adulte d'âge moyen , Liaison aux protéines , Protéolyse , Résultat thérapeutique , Jeune adulte
14.
Curr Opin Nephrol Hypertens ; 26(2): 123-128, 2017 03.
Article de Anglais | MEDLINE | ID: mdl-27977428

RÉSUMÉ

PURPOSE OF REVIEW: The complement system represents one of the more primitive forms of innate immunity. It has increasingly been found to contribute to pathologies in the native and transplanted kidney. We provide a concise review of the physiology of the complement cascade, and discuss current and upcoming complement-based therapies. RECENT FINDINGS: Current agents in clinical use either bind to complement components directly or prevent complement from binding to antibodies affixed to the endothelial surface. These include C1 esterase inhibitors, anti-C5 mAbs, anti-CD20 mAbs, and proteasome inhibitors. Treatment continues to show efficacy in the atypical hemolytic uremic syndrome and antibody-mediated rejection. Promising agents not currently available include CCX168, TP10, AMY-101, factor D inhibitors, coversin, and compstatin. Several new trials are targeting complement inhibition to treat antineutrophilic cystoplasmic antibody (ANCA)-associated vasculitis, C3 glomerulopathy, thrombotic microangiopathy, and IgA nephropathy. New agents for the treatment of the atypical hemolytic uremic syndrome are also in development. SUMMARY: Complement-based therapies are being considered for targeted therapy in the atypical hemolytic uremic syndrome and antibody-mediated rejection, C3 glomerulopathy, and ANCA-associated vasculitis. A few agents are currently in use as orphan drugs. A number of other drugs are in clinical trials and, overall, are showing promising preliminary results.


Sujet(s)
Anticorps monoclonaux/usage thérapeutique , Protéines du système du complément/métabolisme , Dérivés de l'aniline/usage thérapeutique , Vascularites associées aux anticorps anti-cytoplasme des neutrophiles/traitement médicamenteux , Antigènes CD20/immunologie , Syndrome hémolytique et urémique atypique/traitement médicamenteux , Activation du complément/effets des médicaments et des substances chimiques , Complément C5/antagonistes et inhibiteurs , Complément C5/immunologie , Facteur D du complément/antagonistes et inhibiteurs , Glomérulonéphrite à dépôts d'IgA/traitement médicamenteux , Humains , Acides nipécotiques/usage thérapeutique , Peptides cycliques/usage thérapeutique , Inhibiteurs du protéasome/usage thérapeutique , Récepteurs au complément/usage thérapeutique , Microangiopathies thrombotiques/traitement médicamenteux
15.
Nat Chem Biol ; 12(12): 1105-1110, 2016 Dec.
Article de Anglais | MEDLINE | ID: mdl-27775713

RÉSUMÉ

Complement is a key component of the innate immune system, recognizing pathogens and promoting their elimination. Complement component 3 (C3) is the central component of the system. Activation of C3 can be initiated by three distinct routes-the classical, the lectin and the alternative pathways-with the alternative pathway also acting as an amplification loop for the other two pathways. The protease factor D (FD) is essential for this amplification process, which, when dysregulated, predisposes individuals to diverse disorders including age-related macular degeneration and paroxysmal nocturnal hemoglobinuria (PNH). Here we describe the identification of potent and selective small-molecule inhibitors of FD. These inhibitors efficiently block alternative pathway (AP) activation and prevent both C3 deposition onto, and lysis of, PNH erythrocytes. Their oral administration inhibited lipopolysaccharide-induced AP activation in FD-humanized mice. These data demonstrate the feasibility of inhibiting the AP with small-molecule antagonists and support the development of FD inhibitors for the treatment of complement-mediated diseases.


Sujet(s)
Facteur D du complément/antagonistes et inhibiteurs , Voie alterne d'activation du complément/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Bibliothèques de petites molécules/pharmacologie , Animaux , Facteur D du complément/métabolisme , Relation dose-effet des médicaments , Antienzymes/synthèse chimique , Antienzymes/composition chimique , Humains , Lipopolysaccharides/antagonistes et inhibiteurs , Lipopolysaccharides/pharmacologie , Souris , Souris de lignée C57BL , Modèles moléculaires , Structure moléculaire , Bibliothèques de petites molécules/synthèse chimique , Bibliothèques de petites molécules/composition chimique , Relation structure-activité
16.
J Pharmacol Exp Ther ; 355(2): 288-96, 2015 Nov.
Article de Anglais | MEDLINE | ID: mdl-26359312

RÉSUMÉ

Lampalizumab is an antigen-binding fragment of a humanized monoclonal antibody against complement factor D (CFD), a rate-limiting enzyme in the activation and amplification of the alternative complement pathway (ACP), which is in phase III clinical trials for the treatment of geographic atrophy. Understanding of the pharmacokinetics, pharmacodynamics, and biodistribution of lampalizumab following intravitreal administration in the ocular compartments and systemic circulation is limited but crucial for selecting doses that provide optimal efficacy and safety. Here, we sought to construct a semimechanistic and integrated ocular-systemic pharmacokinetic-pharmacodynamic model of lampalizumab in the cynomolgus monkey to provide a quantitative understanding of the ocular and systemic disposition of lampalizumab and CFD inhibition. The model takes into account target-mediated drug disposition, target turnover, and drug distribution across ocular tissues and systemic circulation. Following intravitreal administration, lampalizumab achieves rapid equilibration across ocular tissues. Lampalizumab ocular elimination is relatively slow, with a τ1/2 of approximately 3 days, whereas systemic elimination is rapid, with a τ1/2 of 0.8 hours. Target-independent linear clearance is predominant in the eye, whereas target-mediated clearance is predominant in the systemic circulation. Systemic CFD synthesis was estimated to be high (7.8 mg/day); however, the amount of CFD entering the eye due to influx from the systemic circulation was small (<10%) compared with the lampalizumab dose and is thus expected to have an insignificant impact on the clinical dose-regimen decision. Our findings support the clinical use of intravitreal lampalizumab to achieve significant ocular ACP inhibition while maintaining low systemic exposure and minimal systemic ACP inhibition.


Sujet(s)
Anticorps monoclonaux humanisés/pharmacologie , Facteur D du complément/antagonistes et inhibiteurs , Atrophie géographique/métabolisme , Fragments Fab d'immunoglobuline/pharmacologie , Administration par voie intraveineuse , Animaux , Anticorps monoclonaux humanisés/pharmacocinétique , Anticorps monoclonaux humanisés/usage thérapeutique , Humeur aqueuse/métabolisme , Femelle , Atrophie géographique/traitement médicamenteux , Fragments Fab d'immunoglobuline/usage thérapeutique , Injections intravitréennes , Macaca fascicularis , Mâle , Modèles biologiques , Rétine/métabolisme , Corps vitré/métabolisme
17.
Neurobiol Aging ; 36(10): 2748-56, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26248865

RÉSUMÉ

We here report synthesis for the first time of the acetyl salicylic acid dimer 5,5'-methylenebis(2-acetoxybenzoic acid) (DAS). DAS inhibits aberrant complement activation by selectively blocking factor D of the alternative complement pathway and C9 of the membrane attack complex. We have previously identified aurin tricarboxylic and its oligomers as promising agents in this regard. DAS is much more potent, inhibiting erythrocyte hemolysis by complement-activated serum with an IC50 in the 100-170 nanomolar range. There are numerous conditions where self-damage from the complement system has been implicated in the pathology, including such chronic degenerative diseases of aging as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and age-related macular degeneration. Consequently, there is a high priority for the discovery and development of agents that can successfully treat such conditions. DAS holds considerable promise for being such an agent.


Sujet(s)
Acide acétylsalicylique/analogues et dérivés , Composés benzhydryliques/pharmacologie , Activation du complément/effets des médicaments et des substances chimiques , Facteur D du complément/antagonistes et inhibiteurs , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/étiologie , Animaux , Acide acétylsalicylique/synthèse chimique , Acide acétylsalicylique/pharmacologie , Acide aurintricarboxylique , Composés benzhydryliques/synthèse chimique , Chats , Cellules cultivées , Complément C6/antagonistes et inhibiteurs , Complexe d'attaque membranaire du complément , Voie alterne d'activation du complément , Chiens , Relation dose-effet des médicaments , Découverte de médicament , Érythrocytes/effets des médicaments et des substances chimiques , Hémolyse/effets des médicaments et des substances chimiques , Humains , Dégénérescence maculaire/traitement médicamenteux , Dégénérescence maculaire/étiologie , Thérapie moléculaire ciblée , Rats
18.
Ophthalmic Res ; 54(2): 64-73, 2015.
Article de Anglais | MEDLINE | ID: mdl-26159686

RÉSUMÉ

Age-related macular degeneration (AMD) is a progressive eye disease affecting many elderly individuals. It has a multifactorial pathogenesis and is associated with numerous environmental (e.g. smoking, light and nutrition) and genetic risk factors. A breakthrough in the mechanisms causing AMD is emerging; the involvement of the alternative pathway of the complement system appears to play a pivotal role. This has led to the statement that AMD is a disease caused by a hyperactive complement system, allowing the term 'complementopathy' to define it more precisely. Abundant evidence includes: the identification of drusen components as activators of complement, immunohistochemical data showing the presence of many species of the complement system in the retinal pigment epithelium-Bruch's membrane-choroidocapillary region of AMD eyes, a strong association of AMD with certain genetic complement protein variants, raised complement levels in blood from AMD patients and the preliminary successful treatments of geographic atrophy with complement factor D (FD) inhibitors. FD is the rate-limiting enzyme of the alternative complement pathway, and is produced by adipose tissue. Recent findings suggest that nutrition may play a role in controlling the level of FD in the circulation. Addressing modifiable risk factors such as smoking and nutrition may thus offer opportunities for the prevention of AMD.


Sujet(s)
Facteur D du complément/antagonistes et inhibiteurs , Voie alterne d'activation du complément , Dégénérescence maculaire/immunologie , Vieillissement/physiologie , Facteur D du complément/métabolisme , Humains , Dégénérescence maculaire/génétique , Dégénérescence maculaire/métabolisme , Épithélium pigmentaire de la rétine/métabolisme
19.
J Pharmacol Exp Ther ; 351(3): 527-37, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25232192

RÉSUMÉ

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.


Sujet(s)
Complément C3a/antagonistes et inhibiteurs , Facteur D du complément/antagonistes et inhibiteurs , Fragments Fab d'immunoglobuline/administration et posologie , Dégénérescence maculaire/traitement médicamenteux , Animaux , Bovins , Complément C3a/immunologie , Facteur D du complément/immunologie , Relation dose-effet des médicaments , Femelle , Humains , Fragments Fab d'immunoglobuline/immunologie , Injections intravitréennes , Macaca fascicularis , Dégénérescence maculaire/sang , Dégénérescence maculaire/immunologie , Mâle , Souris , Résultat thérapeutique
20.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 3): 733-43, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24598742

RÉSUMÉ

Human factor D (FD) is a self-inhibited thrombin-like serine proteinase that is critical for amplification of the complement immune response. FD is activated by its substrate through interactions outside the active site. The substrate-binding, or `exosite', region displays a well defined and rigid conformation in FD. In contrast, remarkable flexibility is observed in thrombin and related proteinases, in which Na(+) and ligand binding is implied in allosteric regulation of enzymatic activity through protein dynamics. Here, ensemble refinement (ER) of FD and thrombin crystal structures is used to evaluate structure and dynamics simultaneously. A comparison with previously published NMR data for thrombin supports the ER analysis. The R202A FD variant has enhanced activity towards artificial peptides and simultaneously displays active and inactive conformations of the active site. ER revealed pronounced disorder in the exosite loops for this FD variant, reminiscent of thrombin in the absence of the stabilizing Na(+) ion. These data indicate that FD exhibits conformational dynamics like thrombin, but unlike in thrombin a mechanism has evolved in FD that locks the unbound native state into an ordered inactive conformation via the self-inhibitory loop. Thus, ensemble refinement of X-ray crystal structures may represent an approach alternative to spectroscopy to explore protein dynamics in atomic detail.


Sujet(s)
Simulation de dynamique moléculaire , Domaine catalytique/génétique , Facteur D du complément/antagonistes et inhibiteurs , Facteur D du complément/composition chimique , Facteur D du complément/génétique , Cristallographie aux rayons X , Cellules HEK293 , Humains , Mutation , Conformation des protéines , Protéolyse , Relation structure-activité , Spécificité du substrat/génétique , Thrombine/composition chimique , Thrombine/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE