Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 231
Filtrar
1.
J Immunol ; 208(5): 1248-1258, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35173033

RESUMEN

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare hemolytic disease driven by impaired complement regulation. Mutations in genes encoding the enzymes that build the GPI anchors are causative, with somatic mutations in the PIG-A gene occurring most frequently. As a result, the important membrane-bound complement regulators CD55 and CD59 are missing on the affected hematopoietic stem cells and their progeny, rendering those cells vulnerable to complement attack. Immune escape mechanisms sparing affected PNH stem cells from removal are suspected in the PNH pathogenesis, but molecular mechanisms have not been elucidated. We hypothesized that exuberant complement activity in PNH results in enhanced immune checkpoint interactions, providing a molecular basis for the potential immune escape in PNH. In a series of PNH patients, we found increased expression levels of the checkpoint ligand programmed death-ligand 1 (PD-L1) on granulocytes and monocytes, as well as in the plasma of PNH patients. Mechanistically, we demonstrate that complement activation leading to the decoration of particles/cells with C3- and/or C4-opsonins increased PD-L1 expression on neutrophils and monocytes as shown for different in vitro models of classical or alternative pathway activation. We further establish in vitro that complement inhibition at the level of C3, but not C5, inhibits the alternative pathway-mediated upregulation of PD-L1 and show by means of soluble PD-L1 that this observation translates into the clinical situation when PNH patients are treated with either C3 or C5 inhibitors. Together, the presented data show that the checkpoint ligand PD-L1 is increased in PNH patients, which correlates with proximal complement activation.


Asunto(s)
Antígeno B7-H1/metabolismo , Activación de Complemento/inmunología , Complemento C3/antagonistas & inhibidores , Complemento C5/antagonistas & inhibidores , Hemoglobinuria Paroxística/patología , Antígeno B7-H1/sangre , Antígenos CD55/genética , Antígenos CD59/genética , Complemento C3/inmunología , Complemento C5/inmunología , Granulocitos/metabolismo , Células Madre Hematopoyéticas/citología , Hemoglobinuria Paroxística/inmunología , Humanos , Evasión Inmune/inmunología , Proteínas de la Membrana/genética , Monocitos/metabolismo
2.
Clin Immunol ; 235: 108785, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34147650

RESUMEN

The FDA approval of pegcetacoplan (Empaveli), a PEGylated compstatin-based C3 therapeutic, as a new treatment for paroxysmal nocturnal hemoglobinuria (PNH) marks a milestone in the history of complement drug discovery. Almost 15 years after the approval of the first complement-specific drug for PNH, the anti-C5 antibody eculizumab, a novel class of complement inhibitors with a distinct mechanism of action finally enters the clinic. This landmark decision broadens the spectrum of available complement therapeutics, offering patients with unmet clinical needs or insufficient responses to anti-C5 therapy an alternative treatment option with a broad activity profile. Here we present a brief historical account of this newly approved complement drug, consolidating its approval within the long research record of the compstatin family of peptidic C3 inhibitors.


Asunto(s)
Complemento C3/antagonistas & inhibidores , Hemoglobinuria Paroxística/tratamiento farmacológico , Péptidos Cíclicos/farmacología , Complemento C3/metabolismo , Aprobación de Drogas , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Péptidos Cíclicos/química
3.
Neurobiol Dis ; 160: 105533, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34673149

RESUMEN

Memory impairment is one of the disabling manifestations of multiple sclerosis (MS) possibly present from the early stages of the disease and for which there is no specific treatment. Hippocampal synaptic dysfunction and dendritic loss, associated with microglial activation, can underlie memory deficits, yet the molecular mechanisms driving such hippocampal neurodegeneration need to be elucidated. In early-stage experimental autoimmune encephalomyelitis (EAE) female mice, we assessed the expression level of molecules involved in microglia-neuron interactions within the dentate gyrus and found overexpression of genes of the complement pathway. Compared to sham immunized mice, the central element of the complement cascade, C3, showed the strongest and 10-fold upregulation, while there was no increase of downstream factors such as the terminal component C5. The combination of in situ hybridization with immunofluorescence showed that C3 transcripts were essentially produced by activated microglia. Pharmacological inhibition of C3 activity, by daily administration of rosmarinic acid, was sufficient to prevent early dendritic loss, microglia-mediated phagocytosis of synapses in the dentate gyrus, and memory impairment in EAE mice, while morphological markers of microglial activation were still observed. In line, when EAE was induced in C3 deficient mice (C3KO), dendrites and spines of the dentate gyrus as well as memory abilities were preserved. Altogether, these data highlight the central role of microglial C3 in early hippocampal neurodegeneration and memory impairment in EAE and, therefore, pave the way toward new neuroprotective strategies in MS to prevent cognitive deficit using complement inhibitors.


Asunto(s)
Complemento C3/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Hipocampo/metabolismo , Trastornos de la Memoria/metabolismo , Degeneración Nerviosa/metabolismo , Animales , Cinamatos/farmacología , Complemento C3/antagonistas & inhibidores , Complemento C3/genética , Convertasas de Complemento C3-C5/farmacología , Dendritas/efectos de los fármacos , Dendritas/metabolismo , Depsidos/farmacología , Encefalomielitis Autoinmune Experimental/patología , Hipocampo/efectos de los fármacos , Hipocampo/patología , Trastornos de la Memoria/patología , Ratones , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/metabolismo , Molibdoferredoxina , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Degeneración Nerviosa/patología , Fagocitosis/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Ácido Rosmarínico
4.
Nat Commun ; 12(1): 5456, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34526511

RESUMEN

Sensitized kidney transplant recipients experience high rates of antibody-mediated rejection due to the presence of donor-specific antibodies and immunologic memory. Here we show that transient peri-transplant treatment with the central complement component C3 inhibitor Cp40 significantly prolongs median allograft survival in a sensitized nonhuman primate model. Despite donor-specific antibody levels remaining high, fifty percent of Cp40-treated primates maintain normal kidney function beyond the last day of treatment. Interestingly, presence of antibodies of the IgM class associates with reduced median graft survival (8 vs. 40 days; p = 0.02). Cp40 does not alter lymphocyte depletion by rhesus-specific anti-thymocyte globulin, but inhibits lymphocyte activation and proliferation, resulting in reduced antibody-mediated injury and complement deposition. In summary, Cp40 prevents acute antibody-mediated rejection and prolongs graft survival in primates, and inhibits T and B cell activation and proliferation, suggesting an immunomodulatory effect beyond its direct impact on antibody-mediated injury.


Asunto(s)
Anticuerpos/inmunología , Complemento C3/antagonistas & inhibidores , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/efectos de los fármacos , Trasplante de Riñón/métodos , Macaca mulatta/inmunología , Piridonas/farmacología , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Proliferación Celular/efectos de los fármacos , Complemento C3/inmunología , Complemento C3/metabolismo , Citocinas/sangre , Citocinas/inmunología , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Supervivencia de Injerto/inmunología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Masculino , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Trasplante Homólogo
5.
Drugs ; 81(12): 1423-1430, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34342834

RESUMEN

Pegcetacoplan (Empaveli™) is a PEGylated pentadecapeptide developed by Apellis Pharmaceuticals for the treatment of complement-mediated diseases. It binds to complement component 3 (C3) and its activation fragment C3b, controlling the cleavage of C3 and the generation of the downstream effectors of complement activation and thus both C3b-mediated extravascular haemolysis and terminal complement-mediated intravascular haemolysis. Pegcetacoplan is the first C3-targeted paroxysmal nocturnal haemoglobinuria (PNH) therapy to be approved (in May 2021) in the USA, where it is indicated for the treatment of adults with PNH, including those switching from C5 inhibitor therapy with eculizumab and ravulizumab. A regulatory assessment of pegcetacoplan for the treatment of PNH is currently underway in the EU and Australia. Pegcetacoplan is also being investigated as a therapeutic option in other complement-mediated diseases, including age-related macular degeneration, C3 glomerulopathy and autoimmune haemolytic anaemia. The recommended dosage regimen of pegcetacoplan is 1080 mg twice weekly, administered as a subcutaneous infusion via an infusion pump with a ≥ 20 mL reservoir. This article summarizes the milestones in the development of pegcetacoplan leading to this first approval for the treatment of adults with PNH.


Asunto(s)
Inactivadores del Complemento/uso terapéutico , Hemoglobinuria Paroxística/tratamiento farmacológico , Péptidos Cíclicos/uso terapéutico , Anemia Hemolítica Autoinmune/tratamiento farmacológico , Complemento C3/antagonistas & inhibidores , Complemento C3b/antagonistas & inhibidores , Inactivadores del Complemento/farmacología , Atrofia Geográfica/tratamiento farmacológico , Glomerulonefritis/tratamiento farmacológico , Humanos , Péptidos Cíclicos/farmacología
6.
Acta Neuropathol Commun ; 9(1): 126, 2021 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-34281628

RESUMEN

Activation of the complement system propagates neuroinflammation and brain damage early and chronically after traumatic brain injury (TBI). The complement system is complex and comprises more than 50 components, many of which remain to be characterized in the normal and injured brain. Moreover, complement therapeutic studies have focused on a limited number of histopathological outcomes, which while informative, do not assess the effect of complement inhibition on neuroprotection and inflammation in a comprehensive manner. Using high throughput gene expression technology (NanoString), we simultaneously analyzed complement gene expression profiles with other neuroinflammatory pathway genes at different time points after TBI. We additionally assessed the effects of complement inhibition on neuropathological processes. Analyses of neuroinflammatory genes were performed at days 3, 7, and 28 post injury in male C57BL/6 mice following a controlled cortical impact injury. We also characterized the expression of 59 complement genes at similar time points, and also at 1- and 2-years post injury. Overall, TBI upregulated the expression of markers of astrogliosis, immune cell activation, and cellular stress, and downregulated the expression of neuronal and synaptic markers from day 3 through 28 post injury. Moreover, TBI upregulated gene expression across most complement activation and effector pathways, with an early emphasis on classical pathway genes and with continued upregulation of C2, C3 and C4 expression 2 years post injury. Treatment using the targeted complement inhibitor, CR2-Crry, significantly ameliorated TBI-induced transcriptomic changes at all time points. Nevertheless, some immune and synaptic genes remained dysregulated with CR2-Crry treatment, suggesting adjuvant anti-inflammatory and neurotropic therapy may confer additional neuroprotection. In addition to characterizing complement gene expression in the normal and aging brain, our results demonstrate broad and chronic dysregulation of the complement system after TBI, and strengthen the view that the complement system is an attractive target for TBI therapy.


Asunto(s)
Lesiones Traumáticas del Encéfalo/genética , Lesión Encefálica Crónica/genética , Encéfalo/efectos de los fármacos , Activación de Complemento/genética , Enfermedades Neuroinflamatorias/genética , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/patología , Lesión Encefálica Crónica/inmunología , Lesión Encefálica Crónica/patología , Activación de Complemento/inmunología , Complemento C2/genética , Complemento C2/inmunología , Complemento C3/antagonistas & inhibidores , Complemento C3/genética , Complemento C3/inmunología , Complemento C4/genética , Complemento C4/inmunología , Inactivadores del Complemento/farmacología , Perfilación de la Expresión Génica , Ratones , Enfermedades Neuroinflamatorias/inmunología , Proteínas Recombinantes de Fusión/farmacología , Transcriptoma
7.
Exp Eye Res ; 207: 108583, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33878326

RESUMEN

PURPOSE: Age-related macular degeneration is a slowly progressing disease. Studies have tied disease risk to an overactive complement system. We have previously demonstrated that pathology in two mouse models, the choroidal neovascularization (CNV) model and the smoke-induced ocular pathology (SIOP) model, can be reduced by specifically inhibiting the alternative complement pathway (AP). Here we report on the development of a novel injury-site targeted inhibitor of the alternative pathway, and its characterization in models of retinal degeneration. METHODS: Expression of the danger associated molecular pattern, a modified annexin IV, in injured ARPE-19 cells was confirmed by immunohistochemistry and complementation assays using B4 IgM mAb. Subsequently, a construct was prepared consisting of B4 single chain antibody (scFv) linked to a fragment of the alternative pathway inhibitor, fH (B4-scFv-fH). ARPE-19 cells stably expressing B4-scFv-fH were microencapsulated and administered intravitreally or subcutaneously into C57BL/6 J mice, followed by CNV induction or smoke exposure. Progression of CNV was analyzed using optical coherence tomography, and SIOP using structure-function analyses. B4-scFv-fH targeting and AP specificity was assessed by Western blot and binding experiments. RESULTS: B4-scFv-fH was secreted from encapsulated RPE and inhibited complement in RPE monolayers. B4-scFv-fH capsules reduced CNV and SIOP, and western blotting for breakdown products of C3α, IgM and IgG confirmed a reduction in complement activation and antibody binding in RPE/choroid. CONCLUSIONS: Data supports a role for natural antibodies and neoepitope expression in ocular disease, and describes a novel strategy to target AP-specific complement inhibition to diseased tissue in the eye. PRECIS: AMD risk is tied to an overactive complement system, and ocular injury is reduced by alternative pathway (AP) inhibition in experimental models. We developed a novel inhibitor of the AP that targets an injury-specific danger associated molecular pattern, and characterized it in disease models.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inactivadores del Complemento/uso terapéutico , Vía Alternativa del Complemento/efectos de los fármacos , Modelos Animales de Enfermedad , Inmunoglobulina M/inmunología , Degeneración Retiniana/terapia , Epitelio Pigmentado de la Retina/metabolismo , Animales , Western Blotting , Línea Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Neovascularización Coroidal/diagnóstico por imagen , Neovascularización Coroidal/inmunología , Neovascularización Coroidal/terapia , Complemento C3/antagonistas & inhibidores , Complemento C3/genética , Sistemas de Liberación de Medicamentos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión , Degeneración Retiniana/diagnóstico por imagen , Degeneración Retiniana/inmunología , Tomografía de Coherencia Óptica , Transfección
8.
Ophthalmology ; 128(9): 1325-1336, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33711380

RESUMEN

OBJECTIVES: To evaluate clinical characteristics of eyes in which investigator-determined new-onset exudative age-related macular degeneration (eAMD) developed during the FILLY trial. DESIGN: Post hoc analysis of the phase 2 study of intravitreal pegcetacoplan in geographic atrophy (GA). SUBJECTS: Patients with GA secondary to age-related macular degeneration (AMD), n = 246. INTERVENTION: Either 15 mg intravitreal pegcetacoplan or sham given monthly or every other month for 12 months followed by a 6-month off-treatment period. MAIN OUTCOME MEASURES: Time of new eAMD onset in the study eye, history of eAMD in the fellow eye, presence of double-layer sign (DLS) on structural OCT in the study eye, changes in retinal anatomic features by structural OCT and fluorescein angiography (FA), and changes in visual acuity. RESULTS: Exudation was reported in 26 study eyes across treatment groups over 18 months. Mean time to eAMD diagnosis was 256 days (range, 31-555 days). Overall, a higher proportion of patients with a baseline history of eAMD in the fellow eye (P = 0.016) and a DLS in the study eye (P = 0.0001) demonstrated eAMD. Among study eyes in which eAMD developed, 18 of 26 (69%) had history of fellow-eye eAMD and 19 of 26 (73.1%) had DLS at baseline, compared with 76 of 217 study eyes (35%; P = 0.0007) and 70 of 215 study eyes (32.5%; P < 0.0001), respectively, in which eAMD did not develop. All 21 patients with structural OCT imaging at the time of eAMD diagnosis demonstrated subretinal fluid, intraretinal cysts, or both consistent with exudation. Among 17 patients who underwent FA at eAMD diagnosis, 10 showed detectable macular neovascularization (MNV), all occult lesions. Development of eAMD did not have an appreciable impact on visual acuity, and all patients responded to anti-vascular endothelial growth factor (VEGF) therapy. CONCLUSIONS: Intravitreal pegcetacoplan slowed the rate of GA growth and was associated with an unexpected dose-dependent increased incidence of eAMD with no temporal clustering of onset. Exudative AMD seemed to be associated with baseline eAMD in the contralateral eye and a DLS, suggestive of nonexudative MNV, in the study eye. The safety profile of pegcetacoplan was acceptable to proceed to phase 3 studies without adjustments to enrollment criteria.


Asunto(s)
Complemento C3/antagonistas & inhibidores , Inactivadores del Complemento/efectos adversos , Atrofia Geográfica/tratamiento farmacológico , Péptidos Cíclicos/efectos adversos , Degeneración Macular Húmeda/inducido químicamente , Anciano , Anciano de 80 o más Años , Inactivadores del Complemento/administración & dosificación , Exudados y Transudados , Femenino , Angiografía con Fluoresceína , Atrofia Geográfica/diagnóstico , Atrofia Geográfica/fisiopatología , Humanos , Inyecciones Intravítreas , Masculino , Persona de Mediana Edad , Péptidos Cíclicos/administración & dosificación , Estudios Prospectivos , Método Simple Ciego , Líquido Subretiniano , Factores de Tiempo , Tomografía de Coherencia Óptica , Agudeza Visual/fisiología , Degeneración Macular Húmeda/diagnóstico , Degeneración Macular Húmeda/fisiopatología
9.
Am J Ophthalmol ; 227: 116-124, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33675755

RESUMEN

PURPOSE: To evaluate the effect of select baseline characteristics on geographic atrophy (GA) progression in eyes receiving intravitreal pegcetacoplan or sham. DESIGN: Phase 2 multicenter, randomized, single-masked, sham-controlled trial. METHODS: Patients with GA received 15 mg pegcetacoplan monthly or every other month (EOM), or sham injection monthly or EOM for 12 months. Primary efficacy endpoint was change in GA lesion size (square root) from baseline. Post hoc analysis evaluated the effects of age; gender; lesion size, focality, and location (extrafoveal vs foveal); pseudodrusen status; best-corrected visual acuity (BCVA); and low-luminance deficit (LLD) on GA progression at Month 12. RESULTS: Of 246 randomized patients, 192 with 12-month data were included in this analysis. Overall mean (standard deviation) change in lesion size (mm) was 0.26 (0.17) (P < .01), 0.27 (0.27) (P < .05), and 0.36 (0.21) in the monthly pegcetacoplan (n = 67), EOM pegcetacoplan (n = 58), and sham (n = 67) groups, respectively. In univariate analysis, patients with extrafoveal lesions (P < .001), BCVA ≥20/60 (P = .001), and larger LLD (P = .002) had greater mean changes in lesion size. Multivariate analysis confirmed significant association of extrafoveal lesions (P = .001) and larger LLD (P = .023) with GA progression. Monthly and EOM pegcetacoplan significantly reduced progression (P < .05) when controlling for these risk factors. CONCLUSIONS: Extrafoveal lesions and larger LLD are potential risk factors for GA progression. Pegcetacoplan treatment significantly controlled GA progression even after accounting for these risk factors.


Asunto(s)
Complemento C3/antagonistas & inhibidores , Inactivadores del Complemento/uso terapéutico , Atrofia Geográfica/tratamiento farmacológico , Péptidos Cíclicos/uso terapéutico , Anciano , Anciano de 80 o más Años , Progresión de la Enfermedad , Femenino , Atrofia Geográfica/diagnóstico , Atrofia Geográfica/fisiopatología , Humanos , Inyecciones Intravítreas , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Método Simple Ciego , Resultado del Tratamiento , Trastornos de la Visión/fisiopatología , Agudeza Visual/fisiología
10.
N Engl J Med ; 384(11): 1028-1037, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33730455

RESUMEN

BACKGROUND: Paroxysmal nocturnal hemoglobinuria (PNH) is a rare, acquired disease characterized by chronic complement-mediated hemolysis. C5 inhibition controls intravascular hemolysis in untreated PNH but cannot address extravascular hemolysis. Pegcetacoplan, a pegylated peptide targeting proximal complement protein C3, potentially inhibits both intravascular and extravascular hemolysis. METHODS: We conducted a phase 3 open-label, controlled trial to assess the efficacy and safety of pegcetacoplan as compared with eculizumab in adults with PNH and hemoglobin levels lower than 10.5 g per deciliter despite eculizumab therapy. After a 4-week run-in phase in which all patients received pegcetacoplan plus eculizumab, we randomly assigned patients to subcutaneous pegcetacoplan monotherapy (41 patients) or intravenous eculizumab (39 patients). The primary end point was the mean change in hemoglobin level from baseline to week 16. Additional clinical and hematologic markers of hemolysis and safety were assessed. RESULTS: Pegcetacoplan was superior to eculizumab with respect to the change in hemoglobin level from baseline to week 16, with an adjusted (least squares) mean difference of 3.84 g per deciliter (P<0.001). A total of 35 patients (85%) receiving pegcetacoplan as compared with 6 patients (15%) receiving eculizumab no longer required transfusions. Noninferiority of pegcetacoplan to eculizumab was shown for the change in absolute reticulocyte count but not for the change in lactate dehydrogenase level. Functional Assessment of Chronic Illness Therapy-Fatigue scores improved from baseline in the pegcetacoplan group. The most common adverse events that occurred during treatment in the pegcetacoplan and eculizumab groups were injection site reactions (37% vs. 3%), diarrhea (22% vs. 3%), breakthrough hemolysis (10% vs. 23%), headache (7% vs. 23%), and fatigue (5% vs. 15%). There were no cases of meningitis in either group. CONCLUSIONS: Pegcetacoplan was superior to eculizumab in improving hemoglobin and clinical and hematologic outcomes in patients with PNH by providing broad hemolysis control, including control of intravascular and extravascular hemolysis. (Funded by Apellis Pharmaceuticals; PEGASUS ClinicalTrials.gov, NCT03500549.).


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Complemento C3/antagonistas & inhibidores , Complemento C5/antagonistas & inhibidores , Inactivadores del Complemento/uso terapéutico , Hemoglobinuria Paroxística/tratamiento farmacológico , Péptidos/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales Humanizados/efectos adversos , Inactivadores del Complemento/efectos adversos , Diarrea/inducido químicamente , Quimioterapia Combinada , Transfusión de Eritrocitos , Hemoglobinas/análisis , Hemoglobinuria Paroxística/sangre , Hemoglobinuria Paroxística/terapia , Humanos , Inyecciones Subcutáneas/efectos adversos , Persona de Mediana Edad , Péptidos/efectos adversos , Péptidos Cíclicos
11.
Curr Opin Ophthalmol ; 32(3): 294-300, 2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-33630787

RESUMEN

PURPOSE OF REVIEW: This review describes therapeutic research programs for geographic atrophy (GA) due to age-related macular degeneration (AMD). We highlight clinical trial data from phase I, II, and III studies. RECENT FINDINGS: There are currently no treatments for GA, a form of advanced AMD that causes significant visual morbidity. Currently, therapeutic candidates are being developed to delay further progression of GA or even attempt to reverse some of the damage. The approaches to therapy range from molecular targets to cell transplantation. Studies of these novel treatment approaches have demonstrated varying degrees of success. The progress in understanding the disease pathophysiology as well as clinical trial data is reviewed. SUMMARY: There are promising new treatments to prevent GA progression as well as some that may reverse the disease course.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Inactivadores del Complemento/uso terapéutico , Atrofia Geográfica/terapia , Complemento C3/antagonistas & inhibidores , Complemento C5/antagonistas & inhibidores , Progresión de la Enfermedad , Humanos
12.
Blood ; 137(4): 443-455, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33507296

RESUMEN

Blocking the terminal complement pathway with the C5 inhibitor eculizumab has revolutionized the clinical management of several complement-mediated diseases and has boosted the clinical development of new inhibitors. Data on the C3 inhibitor Compstatin and the C5 inhibitors eculizumab and Coversin reported here demonstrate that C3/C5 convertases function differently from prevailing concepts. Stoichiometric C3 inhibition failed to inhibit C5 activation and lytic activity during strong classical pathway activation, demonstrating a "C3 bypass" activation of C5. We show that, instead of C3b, surface-deposited C4b alone can also recruit and prime C5 for consecutive proteolytic activation. Surface-bound C3b and C4b possess similar affinities for C5. By demonstrating that the fluid phase convertase C3bBb is sufficient to cleave C5 as long as C5 is bound on C3b/C4b-decorated surfaces, we show that surface fixation is necessary only for the C3b/C4b opsonins that prime C5 but not for the catalytic convertase unit C3bBb. Of note, at very high C3b densities, we observed membrane attack complex formation in absence of C5-activating enzymes. This is explained by a conformational activation in which C5 adopts a C5b-like conformation when bound to densely C3b-opsonized surfaces. Stoichiometric C5 inhibitors failed to prevent conformational C5 activation, which explains the clinical phenomenon of residual C5 activity documented for different inhibitors of C5. The new insights into the mechanism of C3/C5 convertases provided here have important implications for the development and therapeutic use of complement inhibitors as well as the interpretation of former clinical and preclinical data.


Asunto(s)
C3 Convertasa de la Vía Alternativa del Complemento/fisiología , Complemento C3/antagonistas & inhibidores , Complemento C4b/fisiología , Complemento C5/antagonistas & inhibidores , Inactivadores del Complemento/farmacología , Vía Clásica del Complemento/efectos de los fármacos , Modelos Inmunológicos , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Membrana Celular/inmunología , Complemento C5/química , Inactivadores del Complemento/uso terapéutico , Complejo de Ataque a Membrana del Sistema Complemento/fisiología , Resistencia a Medicamentos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Modelos Moleculares , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/uso terapéutico , Conformación Proteica
13.
Biomed Pharmacother ; 136: 111234, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33454596

RESUMEN

Complement is an important innate immune defence machinery. Once dysregulated, it is often linked to pathogenesis of diverse autoimmune diseases. Artesunate (ART) is a well-known anti-malarial compound. Recently, ART has been highlighted by its potential therapeutic effects on certain complement-related autoimmune diseases. However, the underlying mechanisms are hitherto unknown. In the present study, we found that ART mediated complement interception as validated by analysis of complement haemolytic assay. In cell-based setup using dying Jurkat cells, ART-mediated complement interception was also confirmed. Further, we newly established an ELISA system selectively allowing complement activation via the classical pathway, the lectin pathway and the alternative pathway, respectively. ELISA analysis revealed that ART dose-dependently inhibited C4 activation, C3 activation and terminal complement complex assembly via the effector pathways. ART was found to blockade C1q, C3 and C5 with a lesser extent to properdin. The interaction of ART with C1q was determined to be mediated via C1q globular head region. FACS analysis using ART-conjugated mesoporous silica particles revealed that ART specifically bound the key therapeutic targets of C1q, C3 and C5 on microparticles. In conclusion, we for the first time report the anti-complement bioactivities of ART and suggest a potential therapeutic benefit of ART in the complement-related human diseases.


Asunto(s)
Artesunato/farmacología , Activación de Complemento/efectos de los fármacos , Inactivadores del Complemento/farmacología , Animales , Complemento C1q/antagonistas & inhibidores , Complemento C1q/metabolismo , Complemento C3/antagonistas & inhibidores , Complemento C3/metabolismo , Complemento C4/antagonistas & inhibidores , Complemento C4/metabolismo , Complemento C5/antagonistas & inhibidores , Complemento C5/metabolismo , Hemólisis/efectos de los fármacos , Humanos , Células Jurkat , Necrosis , Oveja Doméstica
14.
Clin Immunol ; 220: 108598, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32961333

RESUMEN

Growing clinical evidence has implicated complement as a pivotal driver of COVID-19 immunopathology. Deregulated complement activation may fuel cytokine-driven hyper-inflammation, thrombotic microangiopathy and NET-driven immunothrombosis, thereby leading to multi-organ failure. Complement therapeutics have gained traction as candidate drugs for countering the detrimental consequences of SARS-CoV-2 infection. Whether blockade of terminal complement effectors (C5, C5a, or C5aR1) may elicit similar outcomes to upstream intervention at the level of C3 remains debated. Here we compare the efficacy of the C5-targeting monoclonal antibody eculizumab with that of the compstatin-based C3-targeted drug candidate AMY-101 in small independent cohorts of severe COVID-19 patients. Our exploratory study indicates that therapeutic complement inhibition abrogates COVID-19 hyper-inflammation. Both C3 and C5 inhibitors elicit a robust anti-inflammatory response, reflected by a steep decline in C-reactive protein and IL-6 levels, marked lung function improvement, and resolution of SARS-CoV-2-associated acute respiratory distress syndrome (ARDS). C3 inhibition afforded broader therapeutic control in COVID-19 patients by attenuating both C3a and sC5b-9 generation and preventing FB consumption. This broader inhibitory profile was associated with a more robust decline of neutrophil counts, attenuated neutrophil extracellular trap (NET) release, faster serum LDH decline, and more prominent lymphocyte recovery. These early clinical results offer important insights into the differential mechanistic basis and underlying biology of C3 and C5 inhibition in COVID-19 and point to a broader pathogenic involvement of C3-mediated pathways in thromboinflammation. They also support the evaluation of these complement-targeting agents as COVID-19 therapeutics in large prospective trials.


Asunto(s)
Betacoronavirus/patogenicidad , Complemento C3/antagonistas & inhibidores , Complemento C5/antagonistas & inhibidores , Inactivadores del Complemento/uso terapéutico , Infecciones por Coronavirus/tratamiento farmacológico , Factores Inmunológicos/uso terapéutico , Neumonía Viral/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Biomarcadores/sangre , Proteína C-Reactiva/metabolismo , COVID-19 , Estudios de Cohortes , Activación de Complemento/efectos de los fármacos , Complemento C3/genética , Complemento C3/inmunología , Complemento C5/genética , Complemento C5/inmunología , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Trampas Extracelulares/efectos de los fármacos , Femenino , Expresión Génica , Humanos , Interleucina-6/metabolismo , Masculino , Persona de Mediana Edad , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/virología , Pandemias , Péptidos Cíclicos/uso terapéutico , Neumonía Viral/complicaciones , Neumonía Viral/inmunología , Neumonía Viral/virología , Síndrome de Dificultad Respiratoria/complicaciones , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/virología , SARS-CoV-2 , Índice de Severidad de la Enfermedad
15.
J Mol Model ; 26(9): 231, 2020 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-32789582

RESUMEN

The complement system plays a major role in human immunity, but its abnormal activation can have severe pathological impacts. By mimicking a natural mechanism of complement regulation, the small peptide compstatin has proven to be a very promising complement inhibitor. Over the years, several compstatin analogs have been created, with improved inhibitory potency. A recent analog is being developed as a candidate drug against several pathological conditions, including COVID-19. However, the reasons behind its higher potency and increased binding affinity to complement proteins are not fully clear. This computational study highlights the mechanistic properties of several compstatin analogs, thus complementing previous experimental studies. We perform molecular dynamics simulations involving six analogs alone in solution and two complexes with compstatin bound to complement component 3. These simulations reveal that all the analogs we consider, except the original compstatin, naturally adopt a pre-bound conformation in solution. Interestingly, this set of analogs adopting a pre-bound conformation includes analogs that were not known to benefit from this behavior. We also show that the most recent compstatin analog (among those we consider) forms a stronger hydrogen bond network with its complement receptor than an earlier analog.


Asunto(s)
Antivirales/química , Complemento C3/antagonistas & inhibidores , Péptidos Cíclicos/química , Péptidos Cíclicos/metabolismo , Antivirales/metabolismo , COVID-19 , Complemento C3/metabolismo , Infecciones por Coronavirus/tratamiento farmacológico , Humanos , Enlace de Hidrógeno , Simulación de Dinámica Molecular , Pandemias , Neumonía Viral/tratamiento farmacológico , Relación Estructura-Actividad
16.
Brain Res Bull ; 163: 109-119, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32681971

RESUMEN

BACKGROUND: Major depressive disorder (MDD) has been shown to be related to immune inflammation and the complement system. Previous studies have suggested that human umbilical cord mesenchymal stem cells (hUC-MSCs) play an important role in inflammatory diseases. METHODS: hUC-MSCs were administered into chronic unpredictable mild stress model (CUMS) mice through the tail vein once a week for 4 weeks. After the administration of hUC-MSCs, the depression-like and anxiety-like phenotypes, neuronal histopathology, synaptic-related protein expression and inflammatory index of the mice were assessed. Microglial M1/M2 polarization and the expression of C3a in astrocytes and C3aR in microglia was detected by immunofluorescence co-localization. Then, CUMS mice were injected with a C3aR antagonist, and the expression of C3a and C3aR and microglial polarization were observed. RESULTS: Based on the sucrose preference and tail suspension tests, hUC-MSCs ameliorated the depression-like behaviors of CUMS mice. Additionally, the anxiety-like behaviors of CUMS mice in the open-field and plus-maze tests were improved after the administration of hUC-MSCs. hUC-MSCs altered microglia polarization by alleviating complement C3a-C3aR signaling activation, which decreased pro-inflammatory factor levels and increased anti-inflammatory factor levels, alleviating neuronal damage and synaptic deficits. CONCLUSION: hUC-MSCs have therapeutic effects on anxiety-like and depressive-like phenotypes caused by CUMS. They can alter the polarization of microglia by inhibiting C3a-C3aR signaling to reduce neuroinflammation.


Asunto(s)
Astrocitos/metabolismo , Complemento C3/metabolismo , Trastorno Depresivo Mayor/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Microglía/metabolismo , Estrés Psicológico/metabolismo , Cordón Umbilical/trasplante , Animales , Enfermedad Crónica , Complemento C3/antagonistas & inhibidores , Trastorno Depresivo Mayor/psicología , Trastorno Depresivo Mayor/terapia , Humanos , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos ICR , Estrés Psicológico/psicología , Estrés Psicológico/terapia , Cordón Umbilical/citología
17.
Clin Immunol ; 217: 108493, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32526273

Asunto(s)
Complejo Antígeno-Anticuerpo/biosíntesis , Betacoronavirus/patogenicidad , Infecciones por Coronavirus/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Enfermedades del Complejo Inmune/inmunología , Neumonía Viral/inmunología , Síndrome Respiratorio Agudo Grave/inmunología , Vasculitis/inmunología , Anticuerpos Antivirales/biosíntesis , Complejo Antígeno-Anticuerpo/efectos de los fármacos , Betacoronavirus/inmunología , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/inmunología , Vasos Sanguíneos/patología , Vasos Sanguíneos/virología , COVID-19 , Complemento C3/antagonistas & inhibidores , Complemento C3/biosíntesis , Inactivadores del Complemento/uso terapéutico , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/complicaciones , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/virología , Humanos , Enfermedades del Complejo Inmune/complicaciones , Enfermedades del Complejo Inmune/tratamiento farmacológico , Enfermedades del Complejo Inmune/virología , Inmunidad Humoral/efectos de los fármacos , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Interleucina-6/antagonistas & inhibidores , Interleucina-6/biosíntesis , Pandemias , Neumonía Viral/complicaciones , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-1/biosíntesis , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/complicaciones , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/virología , Índice de Severidad de la Enfermedad , Vasculitis/complicaciones , Vasculitis/tratamiento farmacológico , Vasculitis/virología
18.
Bioconjug Chem ; 31(7): 1844-1856, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32598839

RESUMEN

Complement is one of the critical branches of innate immunity that determines the recognition of engineered nanoparticles by immune cells. Antibody-targeted iron oxide nanoparticles are a popular platform for magnetic separations, in vitro diagnostics, and molecular imaging. We used 60 nm cross-linked iron oxide nanoworms (CLIO NWs) modified with antibodies against Her2/neu and EpCAM, which are common markers of blood-borne cancer cells, to understand the role of complement in the selectivity of targeting of tumor cells in whole blood. CLIO NWs showed highly efficient targeting and magnetic isolation of tumor cells spiked in lepirudin-anticoagulated blood, but specificity was low due to high uptake by neutrophils, monocytes, and lymphocytes. Complement C3 opsonization in plasma was predominantly via the alternative pathway regardless of the presence of antibody, PEG, or fluorescent tag, but was higher for antibody-conjugated CLIO NWs. Addition of various soluble inhibitors of complement convertase (compstatin, soluble CD35, and soluble CD55) to whole human blood blocked up to 99% of the uptake of targeted CLIO NWs by leukocytes, which resulted in a more selective magnetic isolation of tumor cells. Using well-characterized nanomaterials, we demonstrate here that complement therapeutics can be used to improve targeting selectivity.


Asunto(s)
Complemento C3/metabolismo , Inactivadores del Complemento/farmacología , Linfocitos/metabolismo , Monocitos/metabolismo , Nanopartículas/química , Neutrófilos/metabolismo , Transporte Biológico , Complemento C3/antagonistas & inhibidores , Sistemas de Liberación de Medicamentos , Colorantes Fluorescentes/química , Humanos
19.
Clin Immunol ; 215: 108450, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32360516

RESUMEN

Acute respiratory distress syndrome (ARDS) is a devastating clinical manifestation of COVID-19 pneumonia and is mainly based on an immune-driven pathology. Mounting evidence suggests that COVID-19 is fueled by a maladaptive host inflammatory response that involves excessive activation of innate immune pathways. While a "cytokine storm" involving IL-6 and other cytokines has been documented, complement C3 activation has been implicated as an initial effector mechanism that exacerbates lung injury in preclinical models of SARS-CoV infection. C3-targeted intervention may provide broader therapeutic control of complement-mediated inflammatory damage in COVID-19 patients. Herein, we report the clinical course of a patient with severe ARDS due to COVID-19 pneumonia who was safely and successfully treated with the compstatin-based complement C3 inhibitor AMY-101.


Asunto(s)
Betacoronavirus/efectos de los fármacos , Activación de Complemento/efectos de los fármacos , Complemento C3/antagonistas & inhibidores , Inactivadores del Complemento/uso terapéutico , Infecciones por Coronavirus/tratamiento farmacológico , Péptidos Cíclicos/uso terapéutico , Neumonía Viral/tratamiento farmacológico , Anciano , Antivirales/uso terapéutico , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/inmunología , Fibrilación Atrial/patología , Fibrilación Atrial/virología , Betacoronavirus/inmunología , Betacoronavirus/patogenicidad , COVID-19 , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Humanos , Hipercolesterolemia/tratamiento farmacológico , Hipercolesterolemia/inmunología , Hipercolesterolemia/patología , Hipercolesterolemia/virología , Hipertensión/tratamiento farmacológico , Hipertensión/inmunología , Hipertensión/patología , Hipertensión/virología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Masculino , Pandemias , Neumonía Viral/inmunología , Neumonía Viral/patología , Neumonía Viral/virología , SARS-CoV-2 , Resultado del Tratamiento
20.
J Biol Chem ; 295(26): 8746-8758, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32376685

RESUMEN

The complement system is a tightly controlled proteolytic cascade in the innate immune system, which tags intruding pathogens and dying host cells for clearance. An essential protein in this process is complement component C3. Uncontrolled complement activation has been implicated in several human diseases and disorders and has spurred the development of therapeutic approaches that modulate the complement system. Here, using purified proteins and several biochemical assays and surface plasmon resonance, we report that our nanobody, hC3Nb2, inhibits C3 deposition by all complement pathways. We observe that the hC3Nb2 nanobody binds human native C3 and its degradation products with low nanomolar affinity and does not interfere with the endogenous regulation of C3b deposition mediated by Factors H and I. Using negative stain EM analysis and functional assays, we demonstrate that hC3Nb2 inhibits the substrate-convertase interaction by binding to the MG3 and MG4 domains of C3 and C3b. Furthermore, we notice that hC3Nb2 is cross-reactive and inhibits the lectin and alternative pathway in murine serum. We conclude that hC3Nb2 is a potent, general, and versatile inhibitor of the human and murine complement cascades. Its cross-reactivity suggests that this nanobody may be valuable for analysis of complement activation within animal models of both acute and chronic diseases.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Complemento C3/antagonistas & inhibidores , Anticuerpos de Dominio Único/farmacología , Animales , Complemento C3/inmunología , Convertasas de Complemento C3-C5/antagonistas & inhibidores , Convertasas de Complemento C3-C5/inmunología , Hemólisis/efectos de los fármacos , Humanos , Ratones , Modelos Moleculares , Ovinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...