Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
1.
Immunol Rev ; 292(1): 102-119, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31562657

RESUMEN

The B cell activating factor (BAFF) inhibitor, belimumab, is the first biologic drug approved for the treatment of SLE, and exhibits modest, but durable, efficacy in decreasing disease flares and organ damage. BAFF and its homolog APRIL are TNF-like cytokines that support the survival and differentiation of B cells at distinct developmental stages. BAFF is a crucial survival factor for transitional and mature B cells that acts as rheostat for the maturation of low-affinity autoreactive cells. In addition, BAFF augments innate B cell responses via complex interactions with the B cell receptor (BCR) and Toll like receptor (TLR) pathways. In this manner, BAFF impacts autoreactive B cell activation via extrafollicular pathways and fine tunes affinity selection within germinal centers (GC). Finally, BAFF and APRIL support plasma cell survival, with differential impacts on IgM- and IgG-producing populations. Therapeutically, BAFF and combined BAFF/APRIL inhibition delays disease onset in diverse murine lupus strains, although responsiveness to BAFF inhibition is model dependent, in keeping with heterogeneity in clinical responses to belimumab treatment in humans. In this review, we discuss the mechanisms whereby BAFF/APRIL signals promote autoreactive B cell activation, discuss whether altered selection accounts for therapeutic benefits of BAFF inhibition, and address whether new insights into BAFF/APRIL family complexity can be exploited to improve human lupus treatments.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor Activador de Células B/antagonistas & inhibidores , Tolerancia Inmunológica/efectos de los fármacos , Lupus Eritematoso Sistémico/tratamiento farmacológico , Animales , Factor Activador de Células B/inmunología , Factor Activador de Células B/metabolismo , Humanos , Tolerancia Inmunológica/inmunología , Inmunosupresores/uso terapéutico , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Receptores de Antígenos de Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
2.
Eur J Immunol ; 51(9): 2225-2236, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34146342

RESUMEN

Polymorphisms in TACI, a BAFF family cytokine receptor, are linked to diverse human immune disorders including common variable immunodeficiency (CVID) and systemic lupus erythematosus (SLE). Functional studies of individual variants show modest impacts on surface TACI expression and/or downstream signal transduction, indicating that relatively subtle variation in TACI activity can impact human B-cell biology. However, significant complexity underlies TACI biology, including both positive and negative regulation of physiologic and pathogenic B-cell responses. To model these contradictory events, we compared the functional impact of TACI deletion on separate models of murine SLE driven by T cell-independent and -dependent breaks in B-cell tolerance. First, we studied whether reduced surface TACI expression was sufficient to protect against progressive BAFF-mediated systemic autoimmunity. Strikingly, despite a relatively modest impact on surface TACI levels, TACI haploinsufficiency markedly reduced pathogenic RNA-associated autoantibody titers and conferred long-term protection from BAFF-driven lupus nephritis. In contrast, B cell-intrinsic TACI deletion exerted a limited impact of autoantibody generation in murine lupus characterized by spontaneous germinal center formation and T cell-dependent humoral autoimmunity. Together, these combined data provide new insights into TACI biology and highlight how TACI signals must be tightly regulated during protective and pathogenic B-cell responses.


Asunto(s)
Autoinmunidad/genética , Factor Activador de Células B/inmunología , Nefritis Lúpica/genética , Nefritis Lúpica/inmunología , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Autoinmunidad/inmunología , Factor Activador de Células B/antagonistas & inhibidores , Factor Activador de Células B/genética , Receptor del Factor Activador de Células B/genética , Linfocitos B/inmunología , Quimera , Femenino , Haploinsuficiencia/genética , Inmunosupresores/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/genética , Transducción de Señal/inmunología , Proteína Activadora Transmembrana y Interactiva del CAML/inmunología
3.
Am J Pathol ; 191(12): 2231-2244, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34509440

RESUMEN

B cell-activating factor (BAFF), part of a tumor necrosis factor family of cytokines, was recently identified as a regulator of atherosclerosis; however, its role in aortic aneurysm has not been determined. Here, the study examined the effect of selective BAFF antagonism using an anti-BAFF antibody (blocks binding of BAFF to receptors BAFF receptor 3, transmembrane activator and CAML interactor, and B-cell maturation antigen) and mBaffR-mFc (blocks binding of BAFF to BAFF receptor 3) on a murine model of abdominal aortic aneurysm (AAA). In a prevention strategy, the antagonists were injected before the induction of AAA, and in an intervention strategy, the antagonists were injected after the induction of AAA. Both strategies attenuated the formation of AAA. In the intervention group, BAFF antagonism depleted most of the mature B-cell subsets in spleen and circulation, leading to enhanced resolution of inflammation in AAA as indicated by decreased infiltration of B cells and proinflammatory macrophages and a reduced number of apoptotic cells. In AAA tissues, B cells and macrophages were found in close contact. In vitro, B cells, irrespective of treatment with BAFF, impaired the efferocytosis activity of macrophages, suggesting a direct innate role of B cells on macrophage function. Altogether, BAFF antagonism affects survival of the mature B cells, promotes resolution of inflammation in the aorta, and attenuates the growth of AAA in mice.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Aneurisma de la Aorta Abdominal/terapia , Factor Activador de Células B/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/farmacología , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/inmunología , Aneurisma de la Aorta Abdominal/patología , Factor Activador de Células B/genética , Factor Activador de Células B/inmunología , Factor Activador de Células B/fisiología , Subgrupos de Linfocitos B/patología , Recuento de Células , Células Cultivadas , Quimiotaxis de Leucocito/fisiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Fragmentos Fc de Inmunoglobulinas/farmacología , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
4.
Rheumatology (Oxford) ; 60(6): 2502-2516, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33512488

RESUMEN

Systemic erythematosus lupus (SLE) is a classic autoimmune disease characterized by multiple autoantibodies and immune-mediated tissue damage. The aetiology of this disease is still unclear. A new drug, belimumab, which acts against the B-lymphocyte stimulator (BLyS), can effectively improve the condition of SLE patients, but it cannot resolve all SLE symptoms. The discovery of novel, precise therapeutic targets is urgently needed. It is well known that abnormal T-cell function is one of the most crucial factors contributing to the pathogenesis of SLE. Protein post-translational modifications (PTMs), including phosphorylation, glycosylation, acetylation, methylation, ubiquitination and SUMOylation have been emphasized for their roles in activating protein activity, maintaining structural stability, regulating protein-protein interactions and mediating signalling pathways, in addition to other biological functions. Summarizing the latest data in this area, this review focuses on the potential roles of diverse PTMs in regulating T-cell function and signalling pathways in SLE pathogenesis, with the goal of identifying new targets for SLE therapy.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Lupus Eritematoso Sistémico/inmunología , Procesamiento Proteico-Postraduccional , Acetilación , Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor Activador de Células B/antagonistas & inhibidores , Proliferación Celular , Glicosilación , Humanos , Inmunosupresores/uso terapéutico , Lupus Eritematoso Sistémico/terapia , Linfopenia/etiología , Metilación , Fosforilación , Transducción de Señal , Sumoilación , Ubiquitinación
5.
Lupus ; 30(11): 1705-1721, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34238087

RESUMEN

Systemic lupus erythematosus (SLE) is a chronic autoimmune inflammatory disease affecting both adults and children. Belimumab is the only biologic approved for SLE, and the first in a class of drugs known as B-lymphocyte stimulator-specific inhibitors. The introduction of intravenous belimumab in 2011 was a major advance, being the first new therapy approved for SLE in over 50 years. As of April 2021, more than 7200 people with SLE have received belimumab in clinical studies, and it is approved in over 75 countries for the treatment of adults with SLE. A subcutaneous, self-injectable belimumab formulation was licensed in 2017 by both the US Food and Drug Administration (FDA) and European Medicines Agency (EMA). Belimumab was then approved for use in children in Europe, the USA and Japan in 2019, and China and Brazil in 2020. Recently, belimumab became the first FDA-approved drug for the treatment of adults with active lupus nephritis (LN), the most-common severe manifestation of SLE.Over the past 10 years, belimumab has established its position as a disease modifier in the SLE treatment paradigms. Robust evidence from randomised clinical studies and observational, real-world studies has demonstrated the tolerability and efficacy of belimumab for reducing disease activity and the risk of new, severe SLE flares. This enables patients to taper their glucocorticoid use, which limits damage accumulation. Significantly more patients with active LN met the criteria for renal responses and were at less risk of a renal-related event or death after receiving belimumab plus standard therapy, compared with standard therapy on top of mandatory steroid reduction. Ongoing clinical studies are evaluating belimumab's effectiveness in various indications beyond SLE. Post-marketing and registry studies are gathering additional data on key areas such as pregnancy outcomes after belimumab exposure and belimumab co-administration with other biologics.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Inmunosupresores , Lupus Eritematoso Sistémico , Adulto , Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor Activador de Células B/antagonistas & inhibidores , Niño , Humanos , Factores Inmunológicos/uso terapéutico , Inmunosupresores/uso terapéutico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Nefritis Lúpica/tratamiento farmacológico , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento
6.
Immunopharmacol Immunotoxicol ; 43(6): 666-673, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34519594

RESUMEN

The pathogenic roles for B cells in autoimmunity include produce pathogenic autoantibodies and modulate immune responses via the production of cytokines and chemokines. The B lymphocyte stimulator BLyS (also known as B-cell-activating factor, BAFF) and APRIL (a proliferation-inducing ligand) are critical factors in the maintenance of the B-cell pool and humoral immunity, namely BLyS modulates the differentiation and maturation of immature B cell, while APRIL modulates the function and survival of long-lived plasma cell, which plays a prominent role in the pathogenesis of autoimmune diseases. Telitacicept is a novel recombinant fusion protein of both the ligand-binding domain of the TACI receptor and the Fc component of human IgG and which is a BLyS/APRIL dual inhibitor. Moreover, telitacicept was developed by Remegen Co., Ltd. in China and is approved to treat systemic lupus erythematosus in China. We review the rationale, clinical evidence, and future perspectives of telitacicept for the treatment of autoimmune disease.HighlightThe B lymphocyte stimulator BLyS (also known as B-cell-activating factor, BAFF) and APRIL (a proliferation-inducing ligand), members of tumor necrosis factor (TNF) family, and which are critical factors in the maintenance of the B-cell pool and humoral immunity.BAFF and APRIL are implicated in the pathogenesis of several human autoimmune diseases with autoreactive B-cell involvement, and targeting both is beneficial for the treatment of autoimmune diseases.Telitacicept is a novel recombinant fusion protein of both the ligand-binding domain of the TACI receptor and the Fc component of human IgG, as a BLyS/APRIL dual inhibitor and which has been approved by National Medical Products Administration (MNPA) for the treatment of patients with SLE in China.With more clinical trials underway, telitacicept may also be approved for the treatment of other autoimmune diseases in the future.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Factor Activador de Células B/antagonistas & inhibidores , Inmunosupresores/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/antagonistas & inhibidores , Animales , Enfermedades Autoinmunes/inmunología , Factor Activador de Células B/inmunología , Ensayos Clínicos como Asunto/métodos , Humanos , Inmunosupresores/farmacología , Proteínas Recombinantes de Fusión/farmacología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología
7.
Int J Mol Sci ; 22(2)2021 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-33467096

RESUMEN

B cell activating factor (BAFF) is a cytokine that plays a role in the survival, proliferation and differentiation of B cells. We proposed to observe the effects of BAFF inhibition on the humoral immune responses of an allosensitized mouse model using HLA.A2 transgenic mice. Wild-type C57BL/6 mice were sensitized with skin allografts from C57BL/6-Tg (HLA-A2.1)1Enge/J mice and were treated with anti-BAFF monoclonal antibody (mAb) (named Sandy-2) or control IgG1 antibody. HLA.A2-specific IgG was reduced in BAFF-inhibited mice compared to the control group (Δ-13.62 vs. Δ27.07, p < 0.05). BAFF inhibition also resulted in increased pre-pro and immature B cell proportions and decreased mature B cells in the bone marrow (p < 0.05 vs. control). In the spleen, an increase in transitional B cells was observed with a significant decrease in marginal and follicular B cells (p < 0.05 vs. control). There was no significant difference in the proportions of long-lived plasma and memory B cells. Microarray analysis showed that 19 gene probes were significantly up- (>2-fold, p < 0.05) or down-regulated (≤2-fold, p < 0.05) in the BAFF-inhibited group. BAFF inhibition successfully reduced alloimmune responses through the reduction in alloantibody production and suppression of B cell differentiation and maturation. Our data suggest that BAFF suppression may serve as a useful target in desensitization therapy.


Asunto(s)
Factor Activador de Células B/antagonistas & inhibidores , Antígeno HLA-A2/inmunología , Inmunización , Aloinjertos/inmunología , Animales , Anticuerpos/inmunología , Linfocitos B/inmunología , Células de la Médula Ósea/inmunología , Inmunoglobulina G/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Trasplante de Piel/efectos adversos , Bazo/citología , Bazo/inmunología
8.
Ann Rheum Dis ; 79(10): 1340-1348, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32699034

RESUMEN

OBJECTIVES: This ongoing Phase-2, randomised, placebo-controlled, double-blind study evaluated the efficacy, safety and pharmacokinetics of intravenous belimumab in childhood-onset systemic lupus erythematosus (cSLE). METHODS: Patients (5 to 17 years) were randomised to belimumab 10 mg/kg intravenous or placebo every 4 weeks, plus standard SLE therapy. Primary endpoint: SLE Responder Index (SRI4) response rate (Week 52). Key major secondary endpoints: proportion of patients achieving the Paediatric Rheumatology International Trials Organisation/American College of Rheumatology (PRINTO/ACR) response using 50 and '30 alternative' definitions (Week 52), and sustained response (Weeks 44 to 52) by SRI4 and Parent Global Assessment of well-being (Parent-global). Safety and pharmacokinetics were assessed. Study not powered for statistical testing. RESULTS: Ninety-three patients were randomised (belimumab, n=53; placebo, n=40). At Week 52, there were numerically more SRI4 responders with belimumab versus placebo (52.8% vs 43.6%; OR 1.49 (95% CI 0.64 to 3.46)). PRINTO/ACR 30 alternative (52.8% vs 27.5%; OR 2.92 (95% CI 1.19 to 7.17)) and PRINTO/ACR 50 (60.4% vs 35.0%; OR 2.74 (95% CI 1.15 to 6.54)) responses were more frequent with belimumab than placebo, as were sustained responses for SRI4 (belimumab, 43.4%; placebo, 41.0%; OR 1.08 (95% CI 0.46 to 2.52)) and Parent-global (belimumab, 59.1%; placebo, 33.3%; OR 3.49 (95% CI 1.23 to 9.91)). Serious adverse events were reported in 17.0% of belimumab patients and 35.0% of placebo patients; one death occurred (placebo). Week-52, geometric mean (95% CI) belimumab trough concentration was 56.2 (45.2 to 69.8) µg/mL. CONCLUSION: The belimumab intravenous pharmacokinetics and benefit-risk profile in cSLE are consistent with adult belimumab studies and the 10 mg/kg every 4 weeks dose is appropriate. TRIAL REGISTRATION NUMBER: NCT01649765.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Inmunosupresores/uso terapéutico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Administración Intravenosa , Adolescente , Factor Activador de Células B/antagonistas & inhibidores , Niño , Preescolar , Método Doble Ciego , Femenino , Humanos , Masculino , Resultado del Tratamiento
9.
Respir Res ; 21(1): 66, 2020 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-32160903

RESUMEN

BACKGROUND: It has been reported that B cell activating factor belonging to the tumor necrosis factor family (BAFF) expression is increased in chronic obstructive pulmonary disease (COPD). However its role in this chronic inflammatory disease is not fully understood. Previous studies have suggested that BAFF also affects T cell function. We therefore investigated the effects of BAFF on T lymphocytes in COPD. METHODS: BAFF was detected in the cells of sputum and the plasma. Peripheral blood mononuclear cells (PBMCs) were isolated from COPD patients and treated with BAFF or BAFF plus BR3-Fc (BAFF antagonist). The apoptosis of CD4+ cells and CD8+ cells was analyzed by flow cytometry. CD4+ cells and CD8+ cells were isolated from peripheral blood of COPD patients respectively and treated with BAFF or BAFF plus BR3-Fc. Interferon-γ (IFN-γ) and interleukin-4 (IL-4) were detected in the CD4+ cells, and perforin and granzyme B were detected in the CD8+ cells. RESULTS: BAFF expression was increased in the cells of sputum and the plasma from COPD patients compared with control subjects. The plasma BAFF levels were inversely correlated with FEV1 percentage of predicted in patients with COPD. BAFF did not significantly alter the apoptosis of CD4+ cells, however it significantly inhibited the apoptosis of CD8+ cells from COPD patients. BAFF increased IFN-γ expression in the CD4+ cells from COPD patients, while it did not significantly alter the expresson of IL-4 in these cells. BAFF increased the expression of perforin and granzyme B in the CD8+ cells from COPD patients. CONCLUSIONS: Our findings indicate that BAFF may be involved in the inflammatory response in COPD via affecting T lymphocytes, suggesting a possible role of BAFF in the pathogenesis of COPD.


Asunto(s)
Factor Activador de Células B/biosíntesis , Factor Activador de Células B/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Factor Activador de Células B/antagonistas & inhibidores , Células Cultivadas , Femenino , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/patología
10.
Int J Mol Sci ; 21(21)2020 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-33126753

RESUMEN

Intra-renal tertiary lymphoid organs (TLOs) are associated with worsened outcome in kidney transplantation (Ktx). We used an anti-BAFF (B cell activating factor) intervention to investigate whether BAFF is required for TLO formation in a full MHC-mismatch Ktx model in rats. Rats received either therapeutic immunosuppression (no rejection, NR) or subtherapeutic immunosuppression (chronic rejection, CR) and were sacrificed on d56. One group additionally received an anti-BAFF antibody (CR + AB). Intra-renal T (CD3+) and B (CD20+) cells, their proliferation (Ki67+), and IgG+ plasma cells were analyzed by immunofluorescence microscopy. Formation of T and B cell zones and TLOs was assessed. Intra-renal expression of TLO-promoting factors, molecules of T:B crosstalk, and B cell differentiation was analyzed by qPCR. Intra-renal B and T cell zones and TLOs were detected in CR and were associated with elevated intra-renal mRNA expression of TLO-promoting factors, including CXCL13, CCL19, lymphotoxin-ß, and BAFF. Intra-renal plasma cells were also elevated in CR. Anti-BAFF treatment significantly decreased intra-renal B cell zones and TLO, as well as intra-renal B cell-derived TLO-promoting factors and B cell differentiation markers. We conclude that BAFF-dependent intra-renal B cells promote TLO formation and advance local adaptive alloimmune responses in chronic rejection.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Factor Activador de Células B/antagonistas & inhibidores , Linfocitos B/citología , Rechazo de Injerto/tratamiento farmacológico , Riñón/citología , Tejido Linfoide/citología , Linfocitos T/citología , Animales , Factor Activador de Células B/inmunología , Factor Activador de Células B/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Rechazo de Injerto/etiología , Rechazo de Injerto/metabolismo , Rechazo de Injerto/patología , Riñón/efectos de los fármacos , Riñón/inmunología , Riñón/metabolismo , Trasplante de Riñón/efectos adversos , Tejido Linfoide/efectos de los fármacos , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Masculino , Ratas , Ratas Endogámicas BN , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo
11.
J Immunol ; 199(11): 3757-3770, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29055002

RESUMEN

In NOD mice and also likely humans, B lymphocytes play an important role as APC-expanding autoreactive T cell responses ultimately causing type 1 diabetes (T1D). Currently, humans at high future T1D risk can only be identified at late prodromal stages of disease indicated by markers such as insulin autoantibodies. When commenced in already insulin autoantibody+ NOD mice, continuous BAFFR-Fc treatment alone or in combination with anti-CD20 (designated combo therapy) inhibited T1D development. Despite eliciting broader B lymphocyte depletion, continuous combo therapy afforded no greater T1D protection than did BAFFR-Fc alone. As previously observed, late disease stage-initiated anti-CD20 monotherapy did not inhibit T1D, and in this study was additionally found to be associated with development of drug-blocking Abs. Promisingly, NOD mice given transient late disease stage BAFFR-Fc monotherapy were rendered T1D resistant. However, combo treatment abrogated the protective effect of transient BAFFR-Fc monotherapy. NOD mice receiving transient BAFF blockade were characterized by an enrichment of regulatory B lymphocytes that inhibit T1D development through IL-10 production, but this population is sensitive to deletion by anti-CD20 treatment. B lymphocytes from transient BAFFR-Fc-treated mice suppressed T cell proliferation to a greater extent than did those from controls. Proportions of B lymphocytes expressing CD73, an ecto-enzyme operating in a pathway converting proinflammatory ATP to anti-inflammatory adenosine, were also temporarily increased by transient BAFFR-Fc treatment, but not anti-CD20 therapy. These collective studies indicate transient BAFFR-Fc-mediated B lymphocyte depletion elicits long-term T1D protection by enriching regulatory B lymphocytes that are deleted by anti-CD20 cotherapy.


Asunto(s)
Factor Activador de Células B/antagonistas & inhibidores , Linfocitos B Reguladores/inmunología , Diabetes Mellitus Tipo 1/inmunología , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Inmunoterapia/métodos , Rituximab/uso terapéutico , Linfocitos T/inmunología , Animales , Receptor del Factor Activador de Células B/genética , Receptor del Factor Activador de Células B/uso terapéutico , Proliferación Celular , Células Cultivadas , Terapia Combinada , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Terapia de Inmunosupresión , Interleucina-10/metabolismo , Depleción Linfocítica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD
12.
Eur J Immunol ; 47(6): 1075-1085, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28383107

RESUMEN

The TNF family cytokines B-cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL) support plasma cell survival. It is known that inhibitors of BAFF only (BAFFR-Fc) or BAFF and APRIL (TACI-Fc) administered early enough in an NZB/NZW F1 mouse model of systemic lupus erythematosus (SLE) ameliorate clinical outcomes, pointing to a pathogenic role of BAFF. In the present study, TACI-Fc administrated at a later stage of disease, after onset of autoimmunity, decreased the number of bone marrow plasma cells and slowed down further formation of autoantibodies. TACI-Fc prevented renal damage during a 12-week treatment period regardless of autoantibody levels, while BAFFR-Fc did not despite a similar BAFF-blocking activity in vivo. TACI-Fc also decreased established plasma cells in a T-dependent hapten/carrier immunization system better than single inhibitors of BAFF or APRIL, and sometimes better than combined single inhibitors with at least equivalent BAFF and APRIL inhibitory activities. These results indicate that TACI-Fc can prevent symptoms of renal damage in a mouse model of SLE when BAFFR-Fc cannot, and point to a plasticity of plasma cells for survival factors. Targeting plasma cells with TACI-Fc might be beneficial to prevent autoantibody-mediated damages in SLE.


Asunto(s)
Modelos Animales de Enfermedad , Lupus Eritematoso Sistémico/inmunología , Células Plasmáticas/inmunología , Proteína Activadora Transmembrana y Interactiva del CAML/administración & dosificación , Animales , Autoanticuerpos/biosíntesis , Autoinmunidad , Factor Activador de Células B/antagonistas & inhibidores , Factor Activador de Células B/inmunología , Receptor del Factor Activador de Células B/administración & dosificación , Receptor del Factor Activador de Células B/inmunología , Linfocitos B/inmunología , Citometría de Flujo , Riñón/inmunología , Riñón/patología , Lupus Eritematoso Sistémico/fisiopatología , Lupus Eritematoso Sistémico/terapia , Ratones , Células Plasmáticas/patología , Proteína Activadora Transmembrana y Interactiva del CAML/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/antagonistas & inhibidores , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología
13.
Ann Rheum Dis ; 77(6): 883-889, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29563108

RESUMEN

BACKGROUND: Targeted inhibitors of B-cell activating factor (BAFF) have been evaluated in phase III trials in over 4000 patients with systemic lupus erythematosus (SLE). Post hoc analyses of these studies identify greater treatment effect in patients entering with higher disease activity, greater corticosteroid doses, anti double-stranded DNA (dsDNA) and low complement C3 or C4. OBJECTIVES: To evaluate the efficacy and safety of blisibimod, a BAFF inhibitor, in a population of patients with SLE enriched for high disease activity. METHODS: 442 patients with SLE with antinuclear antibodies or anti-dsDNA and Safety of Estrogen in Lupus Erythematosus National Assessment - Systemic Lupus Erythematosus Disease Activity Index (SELENA-SLEDAI) score ≥10 on standard-of-care medications were randomised to receive weekly subcutaneous blisibimod (200 mg) or placebo. Corticosteroid taper was encouraged from week 8. The primary end point was the week 52 SLE Responder Index-6 (SRI-6). RESULTS: The SRI-6 primary end point was not met. There was a statistically significant steroid-sparing effect, and significantly more blisibimod-treated subjects achieved corticosteroid taper. Increased blisibimod treatment effect on SRI-6 was observed in subjects who achieved a concomitant decrease in corticosteroid dose from baseline. In subjects with baseline urinary protein:creatinine ratio (UPCR) ≥56.5 mg/mmol, significantly higher proportions of blisibimod subjects achieved >50% reduction in UPCR and/or UPCR <56.5 mg/mmol. Reductions in SLE autoantibodies and B cells, and increases in complement C3 and C4 were observed with blisibimod.Blisibimod was well-tolerated. The most common adverse events were upper respiratory tract infection, urinary tract infection, injection site erythema/reaction and diarrhoea. CONCLUSIONS: Although the SRI-6 end point was not met, blisibimod was associated with successful steroid reduction, decreased proteinuria and biomarker responses. TRIAL REGISTRATION NUMBER: NCT01395745.


Asunto(s)
Factor Activador de Células B/antagonistas & inhibidores , Factores Inmunológicos/uso terapéutico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Proteínas Recombinantes de Fusión/uso terapéutico , Adulto , Anticuerpos Antinucleares/sangre , Linfocitos B/inmunología , Biomarcadores/sangre , Método Doble Ciego , Esquema de Medicación , Quimioterapia Combinada , Femenino , Glucocorticoides/administración & dosificación , Glucocorticoides/uso terapéutico , Humanos , Factores Inmunológicos/administración & dosificación , Factores Inmunológicos/efectos adversos , Inyecciones Subcutáneas , Lupus Eritematoso Sistémico/inmunología , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Prednisona/administración & dosificación , Prednisona/uso terapéutico , Calidad de Vida , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/efectos adversos , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Adulto Joven
14.
J Autoimmun ; 94: 16-32, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30219390

RESUMEN

Nowadays, pharmacologic treatments of autoinflammatory diseases are largely palliative rather than curative. Most of them result in non-specific immunosuppression, which can be associated with broad disruption of natural and induced immunity with significant and sometimes serious unwanted injuries. Among the novel strategies that are under development, tools that modulate the immune system to restore normal tolerance mechanisms are central. In these approaches, peptide therapeutics constitute a class of agents that display many physicochemical advantages. Within this class of potent drugs, the phosphopeptide P140 is very promising for treating patients with lupus, and likely also patients with other chronic inflammatory diseases. We discovered that P140 targets autophagy, a finely orchestrated catabolic process, involved in the regulation of inflammation and in the biology of immune cells. In vitro, P140 acts directly on a particular form of autophagy called chaperone-mediated autophagy, which seems to be hyperactivated in certain subsets of lymphocytes in lupus and in other autoinflammatory settings. In lupus, the "correcting" effect of P140 on autophagy results in a weaker signaling of autoreactive T cells, leading to a significant improvement of pathophysiological status of treated mice. These findings also demonstrated ex vivo in human cells, open novel avenues of therapeutic intervention in pathological conditions, in which specific and not general targeting is highly pursued in the context of the new action plans for personalized medicines.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Autofagia/efectos de los fármacos , Factores Inmunológicos/uso terapéutico , Lupus Eritematoso Sistémico/terapia , Terapia Molecular Dirigida/métodos , Fragmentos de Péptidos/uso terapéutico , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Autofagia/genética , Autofagia/inmunología , Factor Activador de Células B/antagonistas & inhibidores , Factor Activador de Células B/genética , Factor Activador de Células B/inmunología , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Subgrupos Linfocitarios/efectos de los fármacos , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/patología , Ratones , Medicina de Precisión
15.
J Autoimmun ; 95: 179-190, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30385081

RESUMEN

Autoimmunity occurs when an adaptive immune response is directed against a self-antigen. As such, autoimmune reactions associated with the production of autoantibodies are common. These autoantibodies may either be pathogenic by inducing the initial damage to self, or exacerbate the reaction secondarily to the initial damage. In both cases, the pathway(s) leading to exposure of the immune system to the self-antigen inducing the production of autoantibodies is largely unknown. The latter is largely complicating the setting of putative prophylactic treatments. As a consequence, one possible way to control these diseases is to eliminate the cells producing antibodies. We will see that this approach is not yet part of any treatment in autoimmunity. Indeed, all the currently available non-specific immunosuppressive treatments do not target directly quiescent antibody-producing plasma cells. However, treatments aimed at depleting precursors of plasma cells, mature B-lymphocytes and/or antigen-experienced B cells not yet fully differentiated into plasma cells, are emerging. Such strategies were recently proven to be highly successful in several autoimmune disorders by two independent ways. The first way is by induction of B-cell cytotoxicity with an antibody directed against the surface antigen CD20. The second way is by antagonism of a key B-cell survival factor, the B-cell activation factor from the TNF superfamily (BAFF). In the present review, we will focus on the current knowledge regarding the role of a molecule related to BAFF, a proliferation-inducing ligand (APRIL), in autoimmune diseases, which acts on antibody-producing plasma cells. We will discuss expectations deriving from APRIL targeting in autoimmune diseases.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedades Autoinmunes/terapia , Autoinmunidad/efectos de los fármacos , Factor Activador de Células B/inmunología , Terapia Molecular Dirigida/métodos , Células Plasmáticas/efectos de los fármacos , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Antígenos CD20/genética , Antígenos CD20/inmunología , Autoanticuerpos , Autoantígenos/genética , Autoantígenos/inmunología , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Factor Activador de Células B/antagonistas & inhibidores , Factor Activador de Células B/genética , Subgrupos de Linfocitos B/efectos de los fármacos , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/patología , Citotoxicidad Inmunológica , Regulación de la Expresión Génica , Humanos , Inmunidad Humoral/efectos de los fármacos , Inmunosupresores/uso terapéutico , Células Plasmáticas/inmunología , Células Plasmáticas/patología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/antagonistas & inhibidores , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
16.
Rheumatology (Oxford) ; 57(1): 125-133, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29045736

RESUMEN

Objective: The SLE Responder Index (SRI) is a composite endpoint used in SLE trials. This investigation examined the clinical trial elements that drive response measured by the SRI. Methods: Analyses are based on data from two phase 3 trials (n = 2262) that evaluated the impact of an anti-B-cell activating factor antibody on disease activity using SRI-5 as the primary endpoint (ClinicalTrials.gov NCT01196091 and NCT01205438). Results: The SRI-5 response rate at week 52 for all patients was 32.8%. Non-response due to a lack of SLEDAI improvement, concomitant medication non-compliance or dropout was 31, 16.5 and 19.1%, respectively. Non-response due to deterioration in BILAG or Physician's Global Assessment after SLEDAI improvement, concomitant medication compliance and trial completion was 0.5%. Disease activity in three SLEDAI organ systems was highly prevalent at baseline: mucocutaneous, 90.6%; musculoskeletal, 82.9%; and immunologic, 71.6%. Disease activity in each of the other organ systems was <11% of patients. Four clinical manifestations were highly prevalent at baseline: arthritis, 82.6%; rash, 69.2%; alopecia, 58.2%; and mucosal ulcer, 32.5%. The combined prevalence of renal, vascular and CNS disease at baseline was 17.6%; these patients had high SRI-5 response rates. Adjustments to corticosteroids were allowed during the first 24 weeks. Increases in corticosteroids above 2.5 mg/day were observed in 16.2% of placebo patients over the first 24 weeks after randomization. Conclusion: The primary drivers of SRI-5 response were SLEDAI improvement, concomitant medication adherence and trial completion. Arthritis, rash, alopecia and mucosal ulcer were the most prevalent clinical manifestations at baseline. Corticosteroid increases and rare, highly weighted disease manifestations in SLEDAI can confound the SRI signal.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Inmunosupresores/uso terapéutico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Corticoesteroides/uso terapéutico , Factor Activador de Células B/antagonistas & inhibidores , Ensayos Clínicos como Asunto , Ensayos Clínicos Fase III como Asunto , Quimioterapia Combinada , Humanos , Lupus Eritematoso Sistémico/fisiopatología , Cumplimiento de la Medicación , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento
17.
Lupus ; 27(12): 1926-1935, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30180771

RESUMEN

Belimumab, a specific inhibitor of the soluble B lymphocyte stimulator (BlyS), is the first biological drug approved by the United States Food and Drug Administration for the treatment of patients with active systemic lupus erythematosus (SLE) refractory to standard therapy. Given that an imbalance between regulatory T cells (Treg) and interleukin (IL)-17A-secreting T cells (Th17) has been reported in various autoimmune disorders, we assessed the frequency of both Treg and Th17 peripheral blood populations before and after belimumab administration in 20 patients with active SLE refractory to standard therapy. After six months of treatment, the mean SELENA-SLEDAI score as well as the mean anti-double-stranded DNA antibody titers were significantly decreased. In addition, we observed a significant increase in Treg percentages and a parallel, significant decrease in Th17 percentages, accompanied by significantly reduced serum levels of IL-21. In vitro studies showed that Treg purified from belimumab-treated patients were fully functional and displayed a suppressor function similar to that of Treg purified from healthy donors. Belimumab can restore Treg/Th17 balance in SLE patients with uncontrolled disease activity, and this results in decreased flare rate and reduced glucocorticoid dosage.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor Activador de Células B/antagonistas & inhibidores , Lupus Eritematoso Sistémico/tratamiento farmacológico , Linfocitos T Reguladores/efectos de los fármacos , Células Th17/efectos de los fármacos , Adulto , Anticuerpos Antinucleares/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Resultado del Tratamiento
18.
J Biol Chem ; 291(38): 19826-34, 2016 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-27451394

RESUMEN

B cell activating factor of the TNF family (BAFF), also known as B lymphocyte stimulator, is a ligand required for the generation and maintenance of B lymphocytes. In this study, the ability of different monoclonal antibodies to recognize, inhibit, or activate mouse BAFF was investigated. One of them, a mouse IgG1 named Sandy-2, prevented the binding of BAFF to all of its receptors, BAFF receptor, transmembrane activator and calcium modulating ligand interactor, and B cell maturation antigen, at a stoichiometric ratio; blocked the activity of mouse BAFF on a variety of cell-based reporter assays; and antagonized the prosurvival action of BAFF on primary mouse B cells in vitro A single administration of Sandy-2 in mice induced B cell depletion within 2 weeks, down to levels close to those observed in BAFF-deficient mice. This depletion could then be maintained with a chronic treatment. Sandy-2 and a previously described rat IgG1 antibody, 5A8, also formed a pair suitable for the sensitive detection of endogenous circulating BAFF by ELISA or using a homogenous assay. Interestingly, 5A8 and Sandy-5 displayed activities opposite to that of Sandy-2 by stimulating recombinant BAFF in vitro and endogenous BAFF in vivo These tools will prove useful for the detection and functional manipulation of endogenous mouse BAFF and provide an alternative to the widely used BAFF receptor-Fc decoy receptor for the specific depletion of BAFF in mice.


Asunto(s)
Anticuerpos/farmacología , Factor Activador de Células B/antagonistas & inhibidores , Linfocitos B/inmunología , Inmunoglobulina G/farmacología , Animales , Anticuerpos/inmunología , Factor Activador de Células B/genética , Factor Activador de Células B/inmunología , Linfocitos B/patología , Supervivencia Celular/efectos de los fármacos , Hiperplasia , Inmunoglobulina G/inmunología , Depleción Linfocítica/métodos , Ratones , Ratones Noqueados
19.
J Intern Med ; 281(1): 52-64, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27307107

RESUMEN

Systemic lupus erythematosus (SLE) has a complex clinical picture, and a number of defects in the immune system have been described in patients with the disease. Most organs can be involved in SLE, and in addition to the typical major organ manifestations (e.g. from kidneys and the central nervous system), early cardiovascular disease is a major determinant of prognosis. Several important findings during the last decade have increased the understanding of the mechanisms behind the disease characteristics and the underlying autoimmune process. Amongst, these are defects in the handling of apoptotic cells, increased expression of type I interferon-regulated genes and activation of autoreactive B cells, with both the type I interferon system and the B lymphocyte stimulator (BLyS) having key roles. In addition, a large number of genes have been identified that contribute to these abnormalities. It has also become clear that certain SLE risk genes are associated with some organ manifestations, such as STAT4 with nephritis and IRF8 with myocardial infarction. Furthermore, environmental factors that can induce SLE or trigger a disease flare have been identified. As a consequence of this increased knowledge, new treatments for SLE have been developed. The most recently approved drug for SLE is belimumab, which blocks BLyS, and several new therapies and therapeutic strategies are in the pipeline for clinical application.


Asunto(s)
Lupus Eritematoso Sistémico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor Activador de Células B/antagonistas & inhibidores , Ambiente , Humanos , Inmunosupresores/uso terapéutico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología
20.
Curr Rheumatol Rep ; 19(3): 10, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28265845

RESUMEN

PURPOSE OF REVIEW: The treatment of systemic lupus erythematosus (SLE) still depends on non-specific immunosuppression. Herein, we review promising targeted therapies that have the potential to change this therapeutic paradigm. RECENT FINDINGS: Besides the FDA-approved B lymphocyte stimulator (BLyS) inhibitor, belimumab, interferon-α represents a promising treatment target, albeit with modest effectiveness primarily in non-renal SLE. Preclinical and early-phase clinical trials using biologics and small molecules targeting B and T cell activation as well as the cross-talk between these cells also show promise. BLyS and interferon targeting show the most promising results in challenging the current treatment status in non-renal SLE.


Asunto(s)
Productos Biológicos/uso terapéutico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor Activador de Células B/antagonistas & inhibidores , Humanos , Inmunosupresores/uso terapéutico , Interferones/uso terapéutico , Terapia Molecular Dirigida/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA