Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
MAbs ; 9(7): 1129-1142, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28758875

RESUMEN

Immunostimulatory receptors belonging to the tumor necrosis factor receptor (TNFR) superfamily are emerging as promising targets for cancer immunotherapies. To optimize the agonism of therapeutic antibodies to these receptors, Fc engineering of antibodies was applied to facilitate the clustering of cell surface TNFRs to activate downstream signaling pathways. One engineering strategy is to identify Fc mutations that facilitate antibody multimerization on the cell surface directly. From the analyses of the crystal packing of IgG1 structures, we identified a novel set of Fc mutations, T437R and K248E, that facilitated antibody multimerization upon binding to antigens on cell surface. In a NF-κB reporter assay, the engineered T437R/K248E mutations could facilitate enhanced agonism of an anti-OX40 antibody without the dependence on FcγRIIB crosslinking. Nonetheless, the presence of cells expressing FcγRIIB could facilitate a boost of the agonism of the engineered antibody with mutations on IgG1 Fc, but not on the silent IgG2σ Fc. The Fc engineered antibody also showed enhanced effector functions, including antibody-dependent cell-meditated cytotoxicity, antibody-dependent cellular phagocytosis, and complement-dependent cytotoxicity, depending on the IgG subtypes. Also, the engineered antibodies showed normal FcRn binding and pharmacokinetic profiles in mice. In summary, this study elucidated a novel Fc engineering approach to promote antibody multimerization on a cell surface, which could enhance agonism and improve effector function for anti-TNFR antibodies as well as other therapeutic antibodies.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoterapia/métodos , Ingeniería de Proteínas/métodos , Receptores OX40/agonistas , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Humanos , Ratones , Mutación
2.
Antibodies (Basel) ; 6(3)2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31548527

RESUMEN

Engineering of fragment crystallizable (Fc) domains of therapeutic immunoglobulin (IgG) antibodies to eliminate their immune effector functions while retaining other Fc characteristics has numerous applications, including blocking antigens on Fc gamma (Fcγ) receptor-expressing immune cells. We previously reported on a human IgG2 variant termed IgG2σ with barely detectable activity in antibody-dependent cellular cytotoxicity, phagocytosis, complement activity, and Fcγ receptor binding assays. Here, we extend that work to IgG1 and IgG4 antibodies, alternative subtypes which may offer advantages over IgG2 antibodies. In several in vitro and in vivo assays, the IgG1σ and IgG4σ variants showed equal or even lower Fc-related activities than the corresponding IgG2σ variant. In particular, IgG1σ and IgG4σ variants demonstrate complete lack of effector function as measured by antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, antibody-dependent cellular phagocytosis, and in vivo T-cell activation. The IgG1σ and IgG4σ variants showed acceptable solubility and stability, and typical human IgG1 pharmacokinetic profiles in human FcRn-transgenic mice and cynomolgus monkeys. In silico T-cell epitope analyses predict a lack of immunogenicity in humans. Finally, crystal structures and simulations of the IgG1σ and IgG4σ Fc domains can explain the lack of Fc-mediated immune functions. These variants show promise for use in those therapeutic antibodies and Fc fusions for which the Fc domain should be immunologically "silent".

3.
J Biol Chem ; 288(43): 30843-54, 2013 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-23986451

RESUMEN

Molecularly engineered antibodies with fit-for-purpose properties will differentiate next generation antibody therapeutics from traditional IgG1 scaffolds. One requirement for engineering the most appropriate properties for a particular therapeutic area is an understanding of the intricacies of the target microenvironment in which the antibody is expected to function. Our group and others have demonstrated that proteases secreted by invasive tumors and pathological microorganisms are capable of cleaving human IgG1, the most commonly adopted isotype among monoclonal antibody therapeutics. Specific cleavage in the lower hinge of IgG1 results in a loss of Fc-mediated cell-killing functions without a concomitant loss of antigen binding capability or circulating antibody half-life. Proteolytic cleavage in the hinge region by tumor-associated or microbial proteases is postulated as a means of evading host immune responses, and antibodies engineered with potent cell-killing functions that are also resistant to hinge proteolysis are of interest. Mutation of the lower hinge region of an IgG1 resulted in protease resistance but also resulted in a profound loss of Fc-mediated cell-killing functions. In the present study, we demonstrate that specific mutations of the CH2 domain in conjunction with lower hinge mutations can restore and sometimes enhance cell-killing functions while still retaining protease resistance. By identifying mutations that can restore either complement- or Fcγ receptor-mediated functions on a protease-resistant scaffold, we were able to generate a novel protease-resistant platform with selective cell-killing functionality.


Asunto(s)
Anticuerpos Monoclonales , Citotoxicidad Celular Dependiente de Anticuerpos , Sitios de Unión de Anticuerpos , Ingeniería de Proteínas , Proteolisis , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Citotoxicidad Celular Dependiente de Anticuerpos/genética , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Sitios de Unión de Anticuerpos/genética , Sitios de Unión de Anticuerpos/inmunología , Línea Celular , Humanos , Inmunoglobulina G , Receptores de IgG/genética , Receptores de IgG/inmunología
4.
MAbs ; 5(3): 397-405, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23549129

RESUMEN

Transgenic mice expressing human neonatal Fc receptor (FcRn) instead of mouse FcRn are available for IgG antibody pharmacokinetic (PK) studies. Given the interest in a rodent model that offers reliable predictions of antibody PK in monkeys and humans, we set out to test whether the PK of IgG antibodies in such mice correlated with the PK of the same antibodies in primates. We began by using a single research antibody to study the influence of: (1) different transgenic mouse lines that differ in FcRn transgene expression; (2) homozygous vs. hemizygous FcRn transgenic mice; (3) the presence vs. absence of coinjected high-dose human intravenous immunoglobulin (IVIG), and (4) the presence vs. absence of coinjected high-dose human serum albumin (HSA). Results of those studies suggested that use of hemizygous Tg32 mice (Tg32 hemi) not treated with IVIG or HSA offered potential as a predictive model for PK in humans. Mouse PK studies were then done under those conditions with a panel of test antibodies whose PK in mice and primates is not significantly affected by target binding, and for which monkey or human PK data were readily available. Results from the studies revealed significant correlations between terminal half-life or clearance values observed in the mice and the corresponding values reported in humans. A significant relationship in clearance values between mice and monkeys was also observed. These correlations suggest that the Tg32 hemi mouse model, which is both convenient and cost-effective, can offer value in predicting antibody half-life and clearance in primates.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/genética , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Inmunoglobulina G/administración & dosificación , Receptores Fc/genética , Virus Sincitiales Respiratorios/inmunología , Animales , Ensayos Clínicos como Asunto , Femenino , Semivida , Haplorrinos , Heterocigoto , Homocigoto , Humanos , Inmunoglobulinas Intravenosas/administración & dosificación , Tasa de Depuración Metabólica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Valor Predictivo de las Pruebas , Albúmina Sérica/administración & dosificación , Proteínas Virales de Fusión/inmunología
5.
Mol Immunol ; 44(7): 1524-34, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17045339

RESUMEN

Although it is now clear that certain Fc glycan structures on immunoglobulin G (IgG) antibodies (Abs) can have a dramatic influence on binding to selected Fcgamma receptors (FcgammaR) and on Fc-mediated immune functions, the effects of all known Fc glycan structures still have not been exhaustively studied. We report that in vitro analyses of pairs of monoclonal human IgG Abs that differ in the amount of sialic acid in their Fc glycans revealed that, for each of the three Ab pairs we examined, higher levels of sialylation were associated with reduced activity in Ab-dependent cellular cytotoxicity (ADCC) assays. This relationship between sialylation and ADCC activity was observed regardless of whether the differences in the extent of sialylation were derived by different Ab production processes, use of a lectin column to separate monoclonal Ab preparations into differentially sialylated fractions, or use of direct in vitro glycoengineering methods to convert a lesser sialylated Ab into a highly sialylated Ab. Subsequent investigations revealed that, depending on the individual Ab and how the differences in sialylation were derived, the lower ADCC potency of the more sialylated variants was apparently due to lower-affinity binding to FcgammaRIIIa on natural killer (NK) cells and/or, more interestingly, lower-affinity binding to cell-surface antigen. Our data provide the first example of an Fc glycan structure impacting antigen binding and suggest that avoiding Fc glycan sialylation can offer another means of optimizing ADCC activity of Abs.


Asunto(s)
Citotoxicidad Inmunológica , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Ácido N-Acetilneuramínico/análisis , Polisacáridos/química , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Antígenos/inmunología , Secuencia de Carbohidratos , Células Cultivadas , Pruebas Inmunológicas de Citotoxicidad , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Inmunoglobulina G/química , Datos de Secuencia Molecular , Ingeniería de Proteínas , Receptores de IgG/inmunología
6.
Cytokine ; 34(1-2): 114-24, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16750378

RESUMEN

Cytokines in plasmid form can act as potent adjuvants when co-administered with DNA vaccines, resulting in an enhanced immune response to the DNA-encoded antigen. This is true of interleukin-18 (IL-18), which has been shown to serve as an adjuvant in conjunction with certain DNA vaccines. To determine if the properties of IL-18 could be optimized for use as a DNA vaccine adjuvant, a model of IL-18/IL-18R binding was developed to identify variants of human IL-18 that were predicted to improve receptor interactions and potentially bioactivity. The linkage of mature IL-18 to a secretion signal sequence provided improved protein expression from mammalian cells and signal peptidase cleavage of this protein produced the authentic N-terminus. The IL-18 variant proteins secreted this way were bioactive, as demonstrated by their ability to induce interferon gamma (IFNgamma) expression by human peripheral blood mononuclear cells (PBMCs) and to bind to IL-18R, as demonstrated by BIAcore analysis. The IL-18 variants were inhibited by IL-18 binding protein (IL-18BP), the soluble inhibitor of IL-18, as measured by neutralization of the IFNgamma response in PBMCs. One variant, V11I/T63A, demonstrated increases both in bioactivity and mammalian cell expression as compared to native IL-18, indicating that this molecule may be particularly well suited for use as a DNA-encoded vaccine adjuvant.


Asunto(s)
Interleucina-18/genética , Ingeniería de Proteínas/métodos , Secuencia de Aminoácidos , Secuencia de Bases , Clonación Molecular , Variación Genética , Humanos , Interferón gamma/genética , Interleucina-18/química , Interleucina-18/metabolismo , Leucocitos Mononucleares/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Plásmidos/metabolismo , Estructura Terciaria de Proteína , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Transfección , Vacunas de ADN/genética
7.
Vaccine ; 24(16): 3340-52, 2006 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-16472547

RESUMEN

MUC1 (mucin 1) is a tumor-associated antigen that is overexpressed in many adenocarcinomas. Active immunotherapy targeting tumors expressing MUC1 could have great treatment value. MUC1 DNA vaccines were evaluated in MUC1 transgenic (MUC1.Tg) mice challenged with MC38/MUC1+ tumor cells. Vaccination with MUC1 plasmid DNA (pMUC1) alone was insufficient to induce tumor protection. However, co-administration of pMUC1 with a plasmid encoding murine interleukin-18 (pmuIL-18) resulted in significant tumor protection and survival after tumor challenge. Protection was durable in the absence of additional vaccination, as demonstrated by continued protection of vaccinated mice following tumor rechallenge. Mice surviving challenges with MC38/MUC1+ cells showed significant protection after challenge with MUC1(-) MC38 tumor cells, suggesting that these mice had developed immune responses to epitopes shared between the tumor cell lines. Antibody-mediated depletion of lymphocyte subsets demonstrated that protection was due largely to CD4+ T cells. This work demonstrates that a naked DNA vaccine can break tolerance to MUC1 and induce an immune response capable of mediating both significant protection from tumor challenge and increased survival.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Interleucina-18/inmunología , Mucinas/inmunología , Neoplasias Experimentales/prevención & control , Vacunas de ADN/inmunología , Adyuvantes Inmunológicos , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias/genética , Secuencia de Bases , Linfocitos T CD4-Positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Femenino , Vectores Genéticos , Interleucina-18/genética , Depleción Linfocítica , Subgrupos Linfocitarios/inmunología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mucina-1 , Mucinas/genética , Neoplasias Experimentales/inmunología , Plásmidos , Análisis de Supervivencia , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética
8.
Vaccine ; 24(3): 244-53, 2006 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-16135392

RESUMEN

DNA vaccines show efficacy in many preclinical models, but these results have not yet translated to consistent clinical efficacy. Co-administration of molecularly encoded adjuvants is one approach that may enable DNA vaccines to achieve enhanced immune response induction in humans. Interleukin-18 (IL-18) is a Th1-type cytokine that has been shown to augment the activity of DNA vaccines in some preclinical models. A prostate-specific antigen (PSA) DNA vaccine was tested in a mouse tumor model system to explore the impact of co-administration of a pIL-18 plasmid. Low doses of the pPSA vaccine were not capable of inducing tumor protection, but when pIL-18 was co-administered, complete tumor protection was observed in all mice. Tumor protection was mediated by both CD4(+) and CD8(+) T cells. Detailed analysis of the immune response in mice immunized with either pPSA or pPSA/pIL-18 demonstrated that pIL-18 skewed the PSA-specific immune response toward Th1. More importantly, stronger CD4(+) and CD8(+) T cell responses developed in the pPSA/pIL-18-immunized mice, with faster kinetics. These results suggest that IL-18 is a powerful adjuvant molecule that can enhance the development of antigen-specific immunity and vaccine efficacy.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Interleucina-18/farmacología , Células TH1/inmunología , Adyuvantes Inmunológicos , Animales , Especificidad de Anticuerpos , Antimetabolitos , Bromodesoxiuridina , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Proliferación Celular , Citocinas/análisis , Citocinas/biosíntesis , Femenino , Inmunoglobulina G/análisis , Inmunoglobulina G/biosíntesis , Cinética , Ratones , Ratones Endogámicos BALB C , Neoplasias/inmunología , Neoplasias/prevención & control , Plásmidos/genética , Estimulación Química , Linfocitos T Citotóxicos/inmunología , Vacunas de ADN/inmunología
9.
Cancer Immunol Immunother ; 54(11): 1082-94, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16047142

RESUMEN

Prostate specific antigen (PSA) is a serum marker that is widely used in the detection and monitoring of prostate cancer. Though PSA is a self-antigen, T cell responses to PSA epitopes have been detected in healthy men and prostate cancer patients, suggesting it may be used as a target for active immunotherapy of prostate cancer. A PSA DNA vaccine (pPSA) was evaluated in mice and monkeys for its ability to induce antigen-specific immune responses. Mice immunized intradermally with pPSA demonstrated strong PSA-specific humoral and cellular immunity. The anti-PSA immune responses were skewed toward Th1, as shown by high IFNgamma and IL-2 production. The immune response was sufficient to protect mice from challenge with PSA-expressing tumor cells. Tumor protection was durable in the absence of additional vaccination, as demonstrated by protection of vaccinated mice from tumor rechallenge. Furthermore, pPSA vaccination induced PSA-specific antibody titers in male cynomolgus monkeys, which express a closely related PSA gene. These results demonstrate that vaccination with pPSA may be able to break tolerance and can induce an immune response that mediates tumor protection.


Asunto(s)
Antígeno Prostático Específico/inmunología , Neoplasias de la Próstata/prevención & control , Células TH1/inmunología , Vacunas de ADN/inmunología , Animales , Anticuerpos/sangre , Femenino , Humanos , Inmunización , Interferón gamma/biosíntesis , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos BALB C , Linfocitos T Citotóxicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...