Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 136(6): 740-748, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32369559

RESUMEN

The bispecific antibody emicizumab is increasingly used for hemophilia A treatment. However, its specificity for human factors IX and X (FIX and FX) has limited its in vivo functional analysis to primate models of acquired hemophilia. Here, we describe a novel mouse model that allows emicizumab function to be examined. Briefly, FVIII-deficient mice received IV emicizumab 24 hours before tail-clip bleeding was performed. A second infusion with human FIX and FX, administered 5 minutes before bleeding, generated consistent levels of emicizumab (0.7-19 mg/dL for 0.5-10 mg/kg doses) and of both FIX and FX (85 and 101 U/dL, respectively, after dosing at 100 U/kg). Plasma from these mice display FVIII-like activity in assays (diluted activated partial thromboplastin time and thrombin generation), similar to human samples containing emicizumab. Emicizumab doses of 1.5 mg/kg and higher significantly reduced blood loss in a tail-clip-bleeding model using FVIII-deficient mice. However, reduction was incomplete compared with mice treated with human FVIII concentrate, and no difference in efficacy between doses was observed. From this model, we deducted FVIII-like activity from emicizumab that corresponded to a dose of 4.5 U of FVIII per kilogram (ie, 9.0 U/dL). Interestingly, combined with a low FVIII dose (5 U/kg), emicizumab provided enough additive activity to allow complete bleeding arrest. This model could be useful for further in vivo analysis of emicizumab.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor IX/administración & dosificación , Factor X/administración & dosificación , Hemofilia A/tratamiento farmacológico , Hemorragia/tratamiento farmacológico , Modelos Animales , Animales , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/inmunología , Quimioterapia Combinada , Factor IX/análisis , Factor IX/inmunología , Factor VIII/administración & dosificación , Factor VIII/análisis , Factor VIII/uso terapéutico , Factor X/análisis , Factor X/inmunología , Factor XIa/farmacología , Femenino , Hemofilia A/sangre , Hemofilia A/complicaciones , Hemofilia A/inmunología , Hemorragia/etiología , Infusiones Intravenosas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tiempo de Tromboplastina Parcial , Cola (estructura animal)/lesiones , Trombina/biosíntesis
2.
Infect Immun ; 87(5)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30782860

RESUMEN

Coagulation and inflammation are interconnected, suggesting that coagulation plays a key role in the inflammatory response to pathogens. A phenome-wide association study (PheWAS) was used to identify clinical phenotypes of patients with a polymorphism in coagulation factor X. Patients with this single nucleotide polymorphism (SNP) were more likely to be hospitalized with hemostatic and infection-related disorders, suggesting that factor X contributes to the immune response to infection. To investigate this, we modeled infections by human pathogens in a mouse model of factor X deficiency. Factor X-deficient mice were protected from systemic Acinetobacter baumannii infection, suggesting that factor X plays a role in the immune response to A. baumannii Factor X deficiency was associated with reduced cytokine and chemokine production and alterations in immune cell population during infection: factor X-deficient mice demonstrated increased abundance of neutrophils, macrophages, and effector T cells. Together, these results suggest that factor X activity is associated with an inefficient immune response and contributes to the pathology of A. baumannii infection.


Asunto(s)
Infecciones por Acinetobacter/inmunología , Infecciones por Acinetobacter/fisiopatología , Acinetobacter baumannii/inmunología , Factor X/genética , Factor X/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Fenotipo , Polimorfismo Genético
3.
Pediatr Blood Cancer ; 66(11): e27886, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31348595

RESUMEN

BACKGROUND: Emicizumab is a bispecific antibody that bridges factor IXa and factor X to restore hemostasis in patients with hemophilia A (HA). Its efficacy and safety have been proven in multicenter trials. However, real world data regarding its use in very young children are currently lacking. Ancillary test results for monitoring emicizumab's hemostatic effect and their clinical correlations are scarce. METHODS: Children with HA and inhibitors treated by emicizumab were prospectively followed at our center. Laboratory follow-up included rotational thromboelastometry (ROTEM) and thrombin generation (TG), prior to and during treatment. RESULTS: Eleven children whose median age was 26 months were treated by emicizumab and followed for a median of 36 weeks. During follow-up, none experienced hemarthrosis or any other spontaneous bleeds. For 7/11 patients, emicizumab prophylaxis was sufficient to maintain hemostasis without additional supplemental therapy. Only 4/11 patients were occasionally treated with recombinant activated FVII for trauma. Two minor surgeries were safely performed without supplemental therapy while another procedure was complicated by major bleeding. TG parameters improved for all patients, correlating with their clinical status. Interestingly, the lowest TG values were obtained for patients experiencing bleeding episodes, while ROTEM parameters in all patients were close to the normal range. CONCLUSIONS: This study confirms the safety and efficacy of emicizumab in reducing bleeds in young children with HA with inhibitors, including infants. However, surgeries warrant caution as emicizumab prophylaxis may not be sufficient for some procedures. TG may more accurately reflect the hemostasis state than ROTEM in pediatric patients treated with emicizumab.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Hemofilia A/tratamiento farmacológico , Hemorragia/prevención & control , Pérdida de Sangre Quirúrgica/prevención & control , Preescolar , Factor IXa/inmunología , Factor X/inmunología , Estudios de Seguimiento , Hemofilia A/sangre , Hemofilia A/complicaciones , Hemorragia/etiología , Humanos , Lactante , Masculino , Estudios Prospectivos , Tromboelastografía , Trombina/análisis , Resultado del Tratamiento
4.
PLoS Pathog ; 11(2): e1004673, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25658827

RESUMEN

Adenoviruses are common pathogens, mostly targeting ocular, gastrointestinal and respiratory cells, but in some cases infection disseminates, presenting in severe clinical outcomes. Upon dissemination and contact with blood, coagulation factor X (FX) interacts directly with the adenovirus type 5 (Ad5) hexon. FX can act as a bridge to bind heparan sulphate proteoglycans, leading to substantial Ad5 hepatocyte uptake. FX "coating" also protects the virus from host IgM and complement-mediated neutralisation. However, the contribution of FX in determining Ad liver transduction whilst simultaneously shielding the virus from immune attack remains unclear. In this study, we demonstrate that the FX protection mechanism is not conserved amongst Ad types, and identify the hexon hypervariable regions (HVR) of Ad5 as the capsid proteins targeted by this host defense pathway. Using genetic and pharmacological approaches, we manipulate Ad5 HVR interactions to interrogate the interplay between viral cell transduction and immune neutralisation. We show that FX and inhibitory serum components can co-compete and virus neutralisation is influenced by both the location and extent of modifications to the Ad5 HVRs. We engineered Ad5-derived HVRs into the rare, native non FX-binding Ad26 to create Ad26.HVR5C. This enabled the virus to interact with FX at high affinity, as quantified by surface plasmon resonance, FX-mediated cell binding and transduction assays. Concomitantly, Ad26.HVR5C was also sensitised to immune attack in the absence of FX, a direct consequence of the engineered HVRs from Ad5. In both immune competent and deficient animals, Ad26.HVR5C hepatic gene transfer was mediated by FX following intravenous delivery. This study gives mechanistic insight into the pivotal role of the Ad5 HVRs in conferring sensitivity to virus neutralisation by IgM and classical complement-mediated attack. Furthermore, through this gain-of-function approach we demonstrate the dual functionality of FX in protecting Ad26.HVR5C against innate immune factors whilst determining liver targeting.


Asunto(s)
Adenovirus Humanos/inmunología , Anticuerpos Antivirales/inmunología , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Factor X/inmunología , Infecciones por Adenovirus Humanos/inmunología , Infecciones por Adenovirus Humanos/prevención & control , Adenovirus Humanos/genética , Animales , Anticuerpos Neutralizantes/inmunología , Antígenos Virales/genética , Antígenos Virales/inmunología , Línea Celular Tumoral , Variación Genética/genética , Vectores Genéticos/genética , Células HEK293 , Células HeLa , Humanos , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Ratones , Ratones Endogámicos C57BL , Resonancia por Plasmón de Superficie , Transducción Genética , Acoplamiento Viral
5.
Gene Ther ; 23(7): 592-6, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27014840

RESUMEN

Intravenous delivery of adenoviruses is the optimal route for many gene therapy applications. Once in the blood, coagulation factor X (FX) binds to the adenovirus capsid and protects the virion from natural antibody and classical complement-mediated neutralisation in mice. However, to date, no studies have examined the relevance of this FX/viral immune protective mechanism in human samples. In this study, we assessed the effects of blocking FX on adenovirus type 5 (Ad5) activity in the presence of human serum. FX prevented human IgM binding directly to the virus. In individual human sera samples (n=25), approximately half of those screened inhibited adenovirus transduction only when the Ad5-FX interaction was blocked, demonstrating that FX protected the virus from neutralising components in a large proportion of human sera. In contrast, the remainder of sera tested had no inhibitory effects on Ad5 transduction and FX armament was not required for effective gene transfer. In human sera in which FX had a protective role, Ad5 induced lower levels of complement activation in the presence of FX. We therefore demonstrate for the first time the importance of Ad-FX protection in human samples and highlight subject variability and species-specific differences as key considerations for adenoviral gene therapy.


Asunto(s)
Adenoviridae/inmunología , Factor X/inmunología , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Adenoviridae/genética , Línea Celular Tumoral , Vectores Genéticos/sangre , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Células HEK293 , Humanos , Inmunoglobulina M/inmunología , Inyecciones Intravenosas/métodos
6.
Blood ; 124(20): 3165-71, 2014 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-25274508

RESUMEN

ACE910 is a humanized anti-factor IXa/X bispecific antibody mimicking the function of factor VIII (FVIII). We previously demonstrated in nonhuman primates that a single IV dose of ACE910 exerted hemostatic activity against hemophilic bleeds artificially induced in muscles and subcutis, and that a subcutaneous (SC) dose of ACE910 showed a 3-week half-life and nearly 100% bioavailability, offering support for effective prophylaxis for hemophilia A by user-friendly SC dosing. However, there was no direct evidence that such SC dosing of ACE910 would prevent spontaneous bleeds occurring in daily life. In this study, we newly established a long-term primate model of acquired hemophilia A by multiple IV injections of an anti-primate FVIII neutralizing antibody engineered in mouse-monkey chimeric form to reduce its antigenicity. The monkeys in the control group exhibited various spontaneous bleeding symptoms as well as continuous prolongation of activated partial thromboplastin time; notably, all exhibited joint bleeds, which are a hallmark of hemophilia. Weekly SC doses of ACE910 (initial 3.97 mg/kg followed by 1 mg/kg) significantly prevented these bleeding symptoms; notably, no joint bleeding symptoms were observed. ACE910 is expected to prevent spontaneous bleeds and joint damage in hemophilia A patients even with weekly SC dosing, although appropriate clinical investigation is required.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor IXa/inmunología , Factor X/inmunología , Hemofilia A/complicaciones , Hemorragia/complicaciones , Hemorragia/prevención & control , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales Humanizados/inmunología , Línea Celular , Modelos Animales de Enfermedad , Hemorragia/patología , Humanos , Articulaciones/efectos de los fármacos , Articulaciones/patología , Macaca fascicularis , Masculino , Ratones
7.
Haemophilia ; 22 Suppl 5: 36-41, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27405674

RESUMEN

The development of inhibitors to factor VIII (FVIII) or factor IX (FIX) remains a major treatment complication encountered in the treatment of haemophilia. Not all patients with even the same severity and genotype develop inhibitors suggesting an underlying mechanism of tolerance against FVIII- or FIX-related immunity. One mechanism may be central tolerance observed in patients in whom the FVIII mutation enables some production of the protein. The other is a peripheral tolerance mechanism which may be evident in patients with null mutation. Recently, recombinant porcine FVIII (rpFVIII, Obixur, OBI-1, BAX801) has been developed for the haemostatic treatment of both congenital haemophilia with inhibitor (CHAWI) and acquired haemophilia A (AHA). In 28 subjects with AHA with life-/limb-threatening bleeding, rpFVIII reduced or stopped bleeding in all patients within 24 h. The cross-reactivity of anti-human FVIII antibodies to rpFVIII remains around 30-50%. Recently, new therapeutics based on the quite novel concepts have been developed and clinical studies are ongoing. These are humanized asymmetric antibody mimicking FVIIIa function by maintaining a suitable interaction between FIXa and FX (Emicizumab, ACE910), and small interfering RNAs (siRNA, ALN-AT3) suppress liver production of AT through post-transcriptional gene silencing and a humanized anti-TFPI monoclonal antibody (Concizumab). Their main advantages are longer half-life, subcutaneous applicability and efficacy irrespective of the presence of inhibitors which will make it easier to initiate more effective treatment especially early childhood.


Asunto(s)
Factor VIII/inmunología , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/biosíntesis , Anticuerpos Monoclonales Humanizados/genética , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Neutralizantes/sangre , Factor VIII/uso terapéutico , Factor X/inmunología , Factor X/metabolismo , Factor Xa/inmunología , Factor Xa/metabolismo , Hemofilia A/tratamiento farmacológico , Hemofilia A/inmunología , Humanos , Tolerancia Inmunológica , Interferencia de ARN
8.
Rinsho Ketsueki ; 56(6): 623-31, 2015 Jun.
Artículo en Japonés | MEDLINE | ID: mdl-26256871

RESUMEN

Unmet needs of current hemophilia A treatment include the requirement for frequent intravenous infusions, inhibitor development, and containment of high medical costs. In order to overcome these issues, we produced FVIII which mimics a bispecific antibody against FIXa/FX. ACE910 demonstrated hemostatic effects on both ongoing and spontaneous joint bleeding in the primate acquired hemophilia A model. Recently, a phase 1 study for PK, PD, and the safety of ACE910 was initiated. The t1/2 was approximately 30 days. There were no severe ACE910 related adverse events. Furthermore, bleeding was remarkably decreased by weekly subcutaneous administration in patients with severe hemophilia A, regardless of whether an inhibitor was used. ACE910 has the remarkable advantages of prophylactic efficacy which can be achieved by convenient subcutaneous administrations at a markedly reduced frequency.


Asunto(s)
Anticuerpos/uso terapéutico , Factor IXa/inmunología , Factor X/inmunología , Hemofilia A/tratamiento farmacológico , Hemofilia B/tratamiento farmacológico , Animales , Ensayos Clínicos como Asunto , Hemofilia A/inmunología , Hemofilia B/inmunología , Humanos
9.
Blood Cells Mol Dis ; 52(2-3): 116-20, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24074948

RESUMEN

Acquired factor X (FX) deficiency unrelated to amyloidosis is a rare disorder in which an anti-FX antibody is infrequently detected. A patient with severe bleeding due to a calcium ion-dependent anti-FX IgG antibody is described. The FX affinity purified IgG bound the light chain of FX, but not FX lacking its γ-carboxyglutamic acid domain, and binding was enhanced >1000-fold in the presence of calcium ions. The antibody also recognized prothrombin and factor VII with about 100-fold and 1000-fold lower affinity. Like a lupus anticoagulant, increasing concentrations of phospholipids in functional assays reduced the inhibitory activity of the antibody. The effect of these properties of the inhibitor on laboratory diagnostic studies is considered.


Asunto(s)
Autoanticuerpos/inmunología , Factor X/inmunología , Anciano , Autoanticuerpos/sangre , Autoanticuerpos/aislamiento & purificación , Autoanticuerpos/metabolismo , Coagulación Sanguínea , Calcio/metabolismo , Factor X/antagonistas & inhibidores , Deficiencia del Factor X/sangre , Deficiencia del Factor X/inmunología , Deficiencia del Factor X/metabolismo , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunoglobulina G/aislamiento & purificación , Inmunoglobulina G/metabolismo , Masculino , Fosfolípidos/sangre , Fosfolípidos/metabolismo
11.
Blood ; 117(12): 3445-52, 2011 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-21252090

RESUMEN

Coagulation factors VII (FVII), IX (FIX), X (FX), and protein C share the same domain organization but display very different plasma half-lives. It is plausible that the half-life is influenced by the activation peptide, differing in length and glycosylation and missing in FVII. To test this hypothesis, the influence of activation peptides on the plasma half-life of human FVII was studied by administering human FVII variants containing activation peptide motifs to mice. Insertion of the activation peptide from FX gave 4-fold longer terminal half-life (5.5 hours vs 1.4 hours for FVII), whereas the activation peptide from FIX and protein C resulted in half-lives of 4.3 and 1.7 hours, respectively. Using FX's activation peptide we identified the N-linked glycans as structural features important for the half-life. The peptide location within the FVII molecule appeared not to be critical because similar prolongation was obtained with the activation peptide inserted immediately before the normal site of activation and at the C-terminus. However, only the latter variant was activatable, yielding full amidolytic activity and reduced proteolytic activity with preserved long half-life. Our data support that activation peptides function as plasma retention signals and constitute a new manner to extend the half-life of FVII(a).


Asunto(s)
Factor VII/química , Factor VII/farmacocinética , Fragmentos de Péptidos/fisiología , Plasma/metabolismo , Secuencias de Aminoácidos , Animales , Factor IX/química , Factor IX/inmunología , Factor IX/metabolismo , Factor IX/farmacocinética , Factor VII/metabolismo , Factor X/química , Factor X/inmunología , Factor X/metabolismo , Factor X/farmacocinética , Semivida , Humanos , Masculino , Ratones , Proteínas Mutantes/química , Proteínas Mutantes/farmacocinética , Oligopéptidos/química , Oligopéptidos/farmacocinética , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/farmacocinética , Procesamiento Proteico-Postraduccional
12.
Biochem Biophys Res Commun ; 421(2): 170-6, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22490664

RESUMEN

Adenovirus is widely used in gene therapy and vaccination as a viral vector, and its hypervariable regions (HVRs) on hexon are the main antigen recognition sites of adenovirus. The modification of this area by genetic engineering will change the antigenic specificity of the virus. In addition, recent studies have demonstrated the importance of coagulation factor X (FX) in adenovirus serotype 5-mediated liver transduction in vivo. The binding site of adenovirus to FX is the HVRs on hexon. By constructing five proteins containing chimeric HVRs from different adenovirus serotypes, we focused on the antigenic specificity and the affinity for FX of these proteins compared with the corresponding viruses. Our data showed that HVR5 and HVR7 had only a part of hexon activity to neutralizing antibodies (NAbs) compared with the complete activity of HVR1-7. Results also demonstrated a differential high-affinity interaction of the HVRs proteins with FX and indicated that HVRs protein had a similar binding ability with corresponding adenovirus serotype. These results highlighted some properties of chimeric HVRs proteins and revealed the influence on the structure and function of hexon proteins and adenovirus resulting from the HVRs.


Asunto(s)
Adenoviridae/inmunología , Proteínas de la Cápside/inmunología , Factor X/inmunología , Adenoviridae/genética , Secuencia de Aminoácidos , Anticuerpos Neutralizantes/inmunología , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Epítopos , Terapia Genética , Células HEK293 , Humanos , Datos de Secuencia Molecular , Plásmidos , Conformación Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Vacunas Virales/genética , Vacunas Virales/inmunología
13.
Haemophilia ; 18(1): 102-7, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21707870

RESUMEN

Bleeding disorders secondary to acquired non-inhibitory antibodies directed against vitamin K-dependent coagulation proteins are rare. In this report, the authors describe a patient with a low grade lymphoma who presented with a fatal acquired bleeding manifestation and abnormal hemostatic studies resulting from deficiencies in both prothrombin and factor X. Patient plasma samples were collected and studied for the presence of an acquired inhibitor. Levels of plasma coagulation proteins were measured using immunoassay. Patient anti-prothrombin immunoglobulin G was isolated and binding to prothrombin, prothrombin F1.2, factors IX and X was evaluated using immunoblots and competition immunoassay. Prolongation in the prothrombin time and activated partial thromboplastin time suggested a factor deficiency in the common pathway of coagulation. Functional and antigenic levels of both prothrombin and factor X were decreased. An IgG subtype-4 antibody was isolated from patient plasma using affinity chromatography on prothrombin-sepharose. This antibody was found to bind to a common metal-ion-dependent conformational epitope found on the γ-carboxyglutamic acid (Gla) domain of prothrombin, factor X and factor IX. This report represents the first description of an acquired bleeding disorder resulting from a unique cross-reactive auto-antibody against a common metal-ion-dependent antigenic structure on the Gla-domain of the vitamin K-dependent proteins.


Asunto(s)
Autoanticuerpos/inmunología , Trastornos de la Coagulación Sanguínea/inmunología , Factor IX/inmunología , Factor X/inmunología , Protrombina/inmunología , Anciano , Trastornos de la Coagulación Sanguínea/tratamiento farmacológico , Factores de Coagulación Sanguínea/uso terapéutico , Resultado Fatal , Humanos , Linfoma/complicaciones , Linfoma/patología , Masculino , Neoplasias Cutáneas/complicaciones , Neoplasias Cutáneas/patología
14.
Thromb Haemost ; 122(3): 320-328, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-33930902

RESUMEN

Coagulation factor X (F10) amplifies the clotting reaction in the middle of the coagulation cascade, and thus F10 deficiency leads to a bleeding tendency. Isolated acquired F10 deficiency is widely recognized in patients with immunoglobulin light-chain amyloidosis or plasma cell dyscrasias. However, its occurrence as an autoimmune disorder is extremely rare. The Japanese Collaborative Research Group has been conducting a nationwide survey on autoimmune coagulation factor deficiencies (AiCFDs) starting in the last decade; we recently identified three patients with autoimmune F10 deficiency (AiF10D). Furthermore, an extensive literature search was performed, confirming 26 AiF10D and 28 possible cases. Our study revealed that AiF10D patients were younger than patients with other AiCFDs; AiF10D patients included children and were predominantly male. AiF10D was confirmed as a severe type of bleeding diathesis, although its mortality rate was not high. As AiF10D patients showed only low F10 inhibitor titers, they were considered to have nonneutralizing anti-F10 autoantibodies rather than their neutralizing counterparts. Accordingly, immunological anti-F10 antibody detection is highly recommended. Hemostatic and immunosuppressive therapies may help arrest bleeding and eliminate anti-F10 antibodies, leading to a high recovery rate. However, further investigation is necessary to understand the basic characteristics and proper management of AiF10D owing to the limited number of patients.


Asunto(s)
Enfermedades Autoinmunes , Deficiencia del Factor X , Factor X/inmunología , Trastornos Hemorrágicos , Enfermedades Autoinmunes/sangre , Enfermedades Autoinmunes/complicaciones , Manejo de la Enfermedad , Deficiencia del Factor X/complicaciones , Deficiencia del Factor X/inmunología , Trastornos Hemorrágicos/etiología , Trastornos Hemorrágicos/terapia , Humanos
15.
Int J Hematol ; 111(1): 20-30, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30350119

RESUMEN

Hemophilia A is a congenital disorder caused by deficiency or malfunction of coagulation factor (F) VIII. While exogenously provided FVIII effectively reduces bleeding complications in many hemophilia A patients, multiple efforts are underway to develop new drugs to meet the needs that conventional FVIII agents do not. We have been long engaged in creating and clinically developing a humanized anti-FIXa/FX asymmetric bispecific IgG antibody with a FVIIIa-cofactor activity. Since this project was born from a creative and unique idea, our group recognized from the first that it would face many difficulties in the course of research including establishment of industrial manufacturability of an asymmetric bispecific IgG antibody. The group actually faced various challenges, but addressed all of them during about 10 years of research, and successfully created the potent humanized bispecific antibody, emicizumab. Emicizumab has showed clinical benefits in the human trials among which the first one was started in 2012, and has been currently approved in US, EU, Japan, and some other countries. It is now expected to improve the quality of life of patients and their families. In this article, we review the course of the research and clinical development of emicizumab, and describe its molecular characteristics.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor IXa/inmunología , Factor VIIIa/metabolismo , Factor X/inmunología , Hemofilia A/tratamiento farmacológico , Animales , Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Coagulación Sanguínea/efectos de los fármacos , Factor VIII/inmunología , Factor VIII/metabolismo , Hemofilia A/inmunología , Humanos , Ingeniería de Proteínas
16.
Int J Hematol ; 112(5): 621-630, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32748217

RESUMEN

Emicizumab shortens activated partial thromboplastin time (aPTT) greater than Factor (F)VIII. Clot waveform analysis triggered by ellagic acid and tissue factor trigger (Elg/TF) provided a useful means of assessing emicizumab activity. Thrombin generation assays (TGA) using this trigger reagent might also overcome the difficulties associated with aPTT by emicizumab. To compare TGA triggered by Elg/TF and other reagents (FXIa, TF) for evaluating emicizumab activity. Emicizumab, FVIII, or FVIII-bypassing agents (BPAs) were incubated with FVIII-deficient plasmas prior to TGA initiated by Elg/TF (0.2 µM/0.5 pM), FXIa (5.21 pM), or TF (PPP-Reagent LOW®). Emicizumab, FVIII, or BPAs increased peak thrombin generation (peak-Th) dose-dependently using Elg/TF-trigger and the other triggers. Low responses were evident with FXIa-trigger and the enhanced effects remained below normal levels with Elg/TF-trigger. Experiments using FVIII with emicizumab demonstrated an additive effect on peak-Th using Elg/TF-trigger, and this effect appeared to be less at FVIII ≥ 40 IU/dl. BPAs with emicizumab appeared to mediate additive effects, although its effects were variable. Parameters of thrombin generation from BPAs and emicizumab with Elg/TF-trigger were improved to normal level compared to low TF-trigger. Elg/TF-TGA could evaluate global coagulation potential during emicizumab prophylaxis including concomitant therapy with FVIII or BPAs.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Pruebas de Coagulación Sanguínea/métodos , Factor VIII/uso terapéutico , Factor X/inmunología , Hemofilia A/tratamiento farmacológico , Hemorragia/prevención & control , Tiempo de Tromboplastina Parcial , Trombina/metabolismo , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Factor VIII/farmacología , Hemofilia A/sangre , Hemofilia A/complicaciones , Hemorragia/etiología , Humanos
17.
Front Immunol ; 11: 476, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32265927

RESUMEN

Replacement therapy with coagulation factor VIII (FVIII) represents the current clinical treatment for patients affected by hemophilia A (HA). This treatment while effective is, however, hampered by the formation of antibodies which inhibit the activity of infused FVIII in up to 30% of treated patients. Immune tolerance induction (ITI) protocols, which envisage frequent infusions of high doses of FVIII to confront this side effect, dramatically increase the already high costs associated to a patient's therapy and are not always effective in all treated patients. Therefore, there are clear unmet needs that must be addressed in order to improve the outcome of these treatments for HA patients. Taking advantage of preclinical mouse models of hemophilia, several strategies have been proposed in recent years to prevent inhibitor formation and eradicate the pre-existing immunity to FVIII inhibitor positive patients. Herein, we will review some of the most promising strategies developed to avoid and eradicate inhibitors, including the use of immunomodulatory drugs or molecules, oral or transplacental delivery as well as cell and gene therapy approaches. The goal is to improve and potentiate the current ITI protocols and eventually make them obsolete.


Asunto(s)
Factor VIII/inmunología , Hemofilia A/tratamiento farmacológico , Tolerancia Inmunológica/efectos de los fármacos , Factores Inmunológicos/uso terapéutico , Isoanticuerpos/inmunología , Traslado Adoptivo , Animales , Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Portadores de Fármacos , Composición de Medicamentos , Evaluación Preclínica de Medicamentos , Factor IXa/inmunología , Factor VIII/administración & dosificación , Factor VIII/uso terapéutico , Factor X/inmunología , Femenino , Terapias Fetales , Terapia Genética , Hemofilia A/inmunología , Hemofilia A/terapia , Humanos , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Inmunoterapia Adoptiva , Isoanticuerpos/biosíntesis , Tejido Linfoide/inmunología , Ratones , Modelos Animales , Células Vegetales , Embarazo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/uso terapéutico , Linfocitos T Reguladores/trasplante
18.
Sci Immunol ; 4(39)2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31541031

RESUMEN

Immune evasion in the tumor microenvironment (TME) is a crucial barrier for effective cancer therapy, and plasticity of innate immune cells may contribute to failures of targeted immunotherapies. Here, we show that rivaroxaban, a direct inhibitor of activated coagulation factor X (FX), promotes antitumor immunity by enhancing infiltration of dendritic cells and cytotoxic T cells at the tumor site. Profiling FX expression in the TME identifies monocytes and macrophages as crucial sources of extravascular FX. By generating mice with immune cells lacking the ability to produce FX, we show that myeloid cell-derived FX plays a pivotal role in promoting tumor immune evasion. In mouse models of cancer, we report that the efficacy of rivaroxaban is comparable with anti-programmed cell death ligand 1 (PD-L1) therapy and that rivaroxaban synergizes with anti-PD-L1 in improving antitumor immunity. Mechanistically, we demonstrate that FXa promotes immune evasion by signaling through protease-activated receptor 2 and that rivaroxaban specifically targets this cell-autonomous signaling pathway to reprogram tumor-associated macrophages. Collectively, our results have uncovered the importance of FX produced in the TME as a regulator of immune cell activation and suggest translational potential of direct oral anticoagulants to remove persisting roadblocks for immunotherapy and provide extravascular benefits in other diseases.


Asunto(s)
Factor X/inmunología , Neoplasias Mamarias Animales/inmunología , Células Mieloides/inmunología , Animales , Femenino , Humanos , Inmunoterapia , Neoplasias Mamarias Animales/terapia , Ratones , Ratones Endogámicos C57BL
19.
Viruses ; 11(7)2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31284434

RESUMEN

Human adenovirus 5 (HAdV-5) is used as a vector in gene therapy clinical trials, hence its interactions with the host immune system have been widely studied. Previous studies have demonstrated that HAdV-5 binds specifically to murine coagulation factor X (mFX), inhibiting IgM and complement-mediated neutralization. Here, we examined the physical binding of immune components to HAdV-5 by nanoparticle tracking analysis, neutralization assays, mass spectrometry analysis and in vivo experiments. We observed that purified mouse Immunoglobulin M (IgM) antibodies bound to HAdV-5 only in the presence of complement components. Active serum components were demonstrated to bind to HAdV-5 in the presence or absence of mFX, indicating that immune molecules and mFX might bind to different sites. Since binding of mFX to HAdV-5 blocks the neutralization cascade, these findings suggested that not all complement-binding sites may be involved in virion neutralization. Furthermore, the data obtained from serum neutralization experiments suggested that immune molecules other than IgM and IgG may trigger activation of the complement cascade in vitro. In vivo experiments were conducted in immunocompetent C57BL/6 or immuno-deficient Rag2-/- mice. HAdV-5T* (a mutant HAdV-5 unable to bind to human or mFX) was neutralized to some extent in both mouse models, suggesting that murine immunoglobulins were not required for neutralization of HAdV-5 in vivo. Liquid Chromatography-Mass Spectrometry (LC-MS/MS) analysis of HAdV-5 and HAdV-5T* after exposure to murine sera showed stable binding of C3 and C4b in the absence of mFX. In summary, these results suggest that HAdV-5 neutralization can be mediated by both the classical and alternative pathways and that, in the absence of immunoglobulins, the complement cascade can be activated by direct binding of C3 to the virion.


Asunto(s)
Adenovirus Humanos/inmunología , Anticuerpos Antivirales/inmunología , Proteínas del Sistema Complemento/inmunología , Inmunoglobulina M/inmunología , Adenovirus Humanos/genética , Animales , Línea Celular , Activación de Complemento , Proteínas de Unión al ADN/deficiencia , Factor X/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Pruebas de Neutralización , Serogrupo , Virión/inmunología
20.
Am J Hematol ; 83(4): 318-20, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17975806

RESUMEN

A patient with spontaneous hemorrhage from multiple body sites was found to have markedly prolonged international normalized ratio (INR) and activated partial thromboplastin times (aPTT) with incomplete correction of aPTT on mixing studies using normal plasma. The cause of this severe hemorrhage was due to a specific factor X inhibitor. No underlying or associated diseases were found. Initial treatment with fresh frozen plasma, vitamin K, and recombinant activated factor VII (rFVIIa) was unsuccessful. However, therapy utilizing plasma exchange with concomitant intravenous immunoglobulin and corticosteroids resulted in a rapid and sustained normalization of factor X levels with a clinical hemostatic response.


Asunto(s)
Deficiencia del Factor X/terapia , Factor X/antagonistas & inhibidores , Trastornos Hemorrágicos/etiología , Inmunoglobulinas Intravenosas/uso terapéutico , Inmunosupresores/uso terapéutico , Intercambio Plasmático , Prednisona/uso terapéutico , Carcinoma Broncogénico/complicaciones , Carcinoma Broncogénico/cirugía , Terapia Combinada , Factor X/inmunología , Deficiencia del Factor X/etiología , Deficiencia del Factor X/inmunología , Humanos , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/cirugía , Enfermedades Linfáticas/complicaciones , Masculino , Persona de Mediana Edad , Prednisona/administración & dosificación , Virosis/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA