Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Immunol Rev ; 273(1): 312-28, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27558343

RESUMEN

Neutrophils play an important role in cancer. This does not only relate to the well-established prognostic value of the presence of neutrophils, either in the blood or in tumor tissue, in the context of cancer progression or for the monitoring of therapy, but also to their active role in the progression of cancer. In the current review, we describe what is known in general about the role of neutrophils in cancer. What is emerging is a complex, rather heterogeneous picture with both pro- and anti-tumorigenic roles, which apparently differs with cancer type and disease stage. Furthermore, we will discuss the well-known role of neutrophils as myeloid-derived suppressor cells (MDSC), and also on the role of neutrophils as important effector cells during antibody therapy in cancer. It is clear that neutrophils contribute substantially to cancer progression in multiple ways, and this includes both direct effects on the cancer cells and indirect effect on the tumor microenvironment. While in many cases neutrophils have been shown to promote tumor progression, for instance by acting as MDSC, there are also protective effects, particularly when antibody immunotherapy is performed. A better understanding of the role of neutrophils is likely to provide opportunities for immunomodulation and for improving the treatment of cancer patients.


Asunto(s)
Inmunoterapia/métodos , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/inmunología , Neutrófilos/inmunología , Microambiente Tumoral , Animales , Anticuerpos/uso terapéutico , Carcinogénesis , Humanos , Inmunomodulación , Neoplasias/terapia
2.
Eur J Immunol ; 48(2): 344-354, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28952147

RESUMEN

The efficacy of cancer therapeutic antibodies varies considerably among patients. Anti-cancer antibodies act through different mechanisms, including antibody-dependent cellular cytotoxicity (ADCC) triggered via Fcγ receptors (FcγR). This phagocyte ADCC can be promoted by interference with CD47-SIRPα interactions, but the magnitude of this enhancement also varies among individuals. Both FcγR and SIRPα display considerable genetic variation, and we investigated whether this explains some of the variability in ADCC. Because of linkage disequilibrium between FcγR variants the interpretation of previous reports suggesting a potential link between FcγR polymorphisms and ADCC has been troublesome. We performed an integrated genetic analysis that enables stratification. ADCC by activated human neutrophils towards Trastuzumab-coated breast cancer cells was predominantly dependent on FcγRIIa. Neutrophils from individuals with the FcγRIIa-131H polymorphic variant displayed significantly higher killing capacity relative to those with FcγRIIa-131R. Furthermore, ADCC was consistently enhanced by targeting CD47-SIRPα interactions, and there were no significant functional differences between the two most prevalent SIRPα polymorphic variants. Thus, neutrophil ADCC capacity is directly related to the FcγRIIa polymorphism, and targeting CD47-SIRPα interactions enhances ADCC independently of FcγR and SIRPα genotype, thereby further suggesting that CD47-SIRPα interference might be a generic strategy for potentiating the efficacy of antibody therapy in cancer.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/genética , Antígenos de Diferenciación/genética , Neoplasias de la Mama/genética , Genotipo , Inmunoterapia/métodos , Neutrófilos/fisiología , Receptores de IgG/genética , Receptores Inmunológicos/genética , Antígenos de Diferenciación/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/inmunología , Antígeno CD47/metabolismo , Línea Celular Tumoral , Femenino , Predisposición Genética a la Enfermedad , Humanos , Desequilibrio de Ligamiento , Polimorfismo Genético , Receptor ErbB-2/inmunología , Receptores de IgG/metabolismo , Receptores Inmunológicos/metabolismo , Trastuzumab/farmacología , Trastuzumab/uso terapéutico , Resultado del Tratamiento
3.
J Immunol ; 199(1): 204-211, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28566370

RESUMEN

Abs of the IgG isotype are glycosylated in their Fc domain at a conserved asparagine at position 297. Removal of the core fucose of this glycan greatly increases the affinity for FcγRIII, resulting in enhanced FcγRIII-mediated effector functions. Normal plasma IgG contains ∼94% fucosylated Abs, but alloantibodies against, for example, Rhesus D (RhD) and platelet Ags frequently have reduced fucosylation that enhances their pathogenicity. The increased FcγRIII-mediated effector functions have been put to use in various afucosylated therapeutic Abs in anticancer treatment. To test the functional consequences of Ab fucosylation, we produced V-gene-matched recombinant anti-RhD IgG Abs of the four different subclasses (IgG1-4) with and without core fucose (i.e., 20% fucose remaining). Binding to all human FcγR types and their functional isoforms was assessed with surface plasmon resonance. All hypofucosylated anti-RhD IgGs of all IgG subclasses indeed showed enhanced binding affinity for isolated FcγRIII isoforms, without affecting binding affinity to other FcγRs. In contrast, when testing hypofucosylated anti-RhD Abs with FcγRIIIa-expressing NK cells, a 12- and 7-fold increased erythrocyte lysis was observed with the IgG1 and IgG3, respectively, but no increase with IgG2 and IgG4 anti-RhD Abs. Notably, none of the hypofucosylated IgGs enhanced effector function of macrophages, which, in contrast to NK cells, express a complex set of FcγRs, including FcγRIIIa. Our data suggest that the beneficial effects of afucosylated biologicals for clinical use can be particularly anticipated when there is a substantial involvement of FcγRIIIa-expressing cells, such as NK cells.


Asunto(s)
Fucosa/química , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Receptores de IgG/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Fucosa/inmunología , Fucosa/metabolismo , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Glicosilación , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/aislamiento & purificación , Inmunoglobulina G/metabolismo , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Unión Proteica , Receptores de IgG/química , Receptores de IgG/genética , Sistema del Grupo Sanguíneo Rh-Hr/inmunología , Resonancia por Plasmón de Superficie
4.
Blood Adv ; 6(7): 2156-2166, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-34942000

RESUMEN

Anti-CD20 antibodies such as rituximab are broadly used to treat B-cell malignancies. These antibodies can induce various effector functions, including immune cell-mediated antibody-dependent cellular cytotoxicity (ADCC). Neutrophils can induce ADCC toward solid cancer cells by trogoptosis, a cytotoxic mechanism known to be dependent on trogocytosis. However, neutrophils seem to be incapable of killing rituximab-opsonized B-cell lymphoma cells. Nevertheless, neutrophils do trogocytose rituximab-opsonized B-cell lymphoma cells, but this only reduces CD20 surface expression and is thought to render tumor cells therapeutically resistant to further rituximab-dependent destruction. Here, we demonstrate that resistance of B-cell lymphoma cells toward neutrophil killing can be overcome by a combination of CD47-SIRPα checkpoint blockade and sodium stibogluconate (SSG), an anti-leishmaniasis drug and documented inhibitor of the tyrosine phosphatase SHP-1. SSG enhanced neutrophil-mediated ADCC of solid tumor cells but enabled trogoptotic killing of B-cell lymphoma cells by turning trogocytosis from a mechanism that contributes to resistance into a cytotoxic anti-cancer mechanism. Tumor cell killing in the presence of SSG required both antibody opsonization of the target cells and disruption of CD47-SIRPα interactions. These results provide a more detailed understanding of the role of neutrophil trogocytosis in antibody-mediated destruction of B cells and clues on how to further optimize antibody therapy of B-cell malignancies.


Asunto(s)
Antígeno CD47 , Neutrófilos , Citotoxicidad Celular Dependiente de Anticuerpos , Gluconato de Sodio Antimonio , Antígeno CD47/metabolismo , Neutrófilos/metabolismo , Rituximab/farmacología , Rituximab/uso terapéutico
5.
Front Immunol ; 12: 594773, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33790888

RESUMEN

Antibody-mediated blood disorders ensue after auto- or alloimmunization against blood cell antigens, resulting in cytopenia. Although the mechanisms of cell destruction are the same as in immunotherapies targeting tumor cells, many factors are still unknown. Antibody titers, for example, often do not strictly correlate with clinical outcome. Previously, we found C-reactive protein (CRP) levels to be elevated in thrombocytopenic patients, correlating with thrombocyte counts, and bleeding severity. Functionally, CRP amplified antibody-mediated phagocytosis of thrombocytes by phagocytes. To investigate whether CRP is a general enhancer of IgG-mediated target cell destruction, we extensively studied the effect of CRP on in vitro IgG-Fc receptor (FcγR)-mediated cell destruction: through respiratory burst, phagocytosis, and cellular cytotoxicity by a variety of effector cells. We now demonstrate that CRP also enhances IgG-mediated effector functions toward opsonized erythrocytes, in particular by activated neutrophils. We performed a first-of-a-kind profiling of CRP binding to all human FcγRs and IgA-Fc receptor I (FcαRI) using a surface plasmon resonance array. CRP bound these receptors with relative affinities of FcγRIa = FcγRIIa/b = FcγRIIIa > FcγRIIIb = FcαRI. Furthermore, FcγR blocking (in particular FcγRIa) abrogated CRP's ability to amplify IgG-mediated neutrophil effector functions toward opsonized erythrocytes. Finally, we observed that CRP also amplified killing of breast-cancer tumor cell line SKBR3 by neutrophils through anti-Her2 (trastuzumab). Altogether, we provide for the first time evidence for the involvement of specific CRP-FcγR interactions in the exacerbation of in vitro IgG-mediated cellular destruction; a trait that should be further evaluated as potential therapeutic target e.g., for tumor eradication.


Asunto(s)
Proteína C-Reactiva/metabolismo , Inmunoglobulina G/inmunología , Receptores Fc/metabolismo , Receptores de IgG/metabolismo , Adulto , Animales , Células Cultivadas , Citofagocitosis/inmunología , Citotoxicidad Inmunológica , Eritrocitos/inmunología , Femenino , Humanos , Inmunoglobulina G/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Modelos Biológicos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Estallido Respiratorio/inmunología , Adulto Joven
6.
Cancer Immunol Res ; 8(1): 120-130, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31690649

RESUMEN

Therapeutic monoclonal antibodies (mAb), directed toward either tumor antigens or inhibitory checkpoints on immune cells, are effective in cancer therapy. Increasing evidence suggests that the therapeutic efficacy of these tumor antigen-targeting mAbs is mediated-at least partially-by myeloid effector cells, which are controlled by the innate immune-checkpoint interaction between CD47 and SIRPα. We and others have previously demonstrated that inhibiting CD47-SIRPα interactions can substantially potentiate antibody-dependent cellular phagocytosis and cytotoxicity of tumor cells by IgG antibodies both in vivo and in vitro IgA antibodies are superior in killing cancer cells by neutrophils compared with IgG antibodies with the same variable regions, but the impact of CD47-SIRPα on IgA-mediated killing has not been investigated. Here, we show that checkpoint inhibition of CD47-SIRPα interactions further enhances destruction of IgA antibody-opsonized cancer cells by human neutrophils. This was shown for multiple tumor types and IgA antibodies against different antigens, i.e., HER2/neu and EGFR. Consequently, combining IgA antibodies against HER2/neu or EGFR with SIRPα inhibition proved to be effective in eradicating cancer cells in vivo In a syngeneic in vivo model, the eradication of cancer cells was predominantly mediated by granulocytes, which were actively recruited to the tumor site by SIRPα blockade. We conclude that IgA-mediated tumor cell destruction can be further enhanced by CD47-SIRPα checkpoint inhibition. These findings provide a basis for targeting CD47-SIRPα interactions in combination with IgA therapeutic antibodies to improve their potential clinical efficacy in tumor patients.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Antígeno CD47/antagonistas & inhibidores , Inmunoglobulina A/inmunología , Neutrófilos/inmunología , Receptores Inmunológicos/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antígenos de Diferenciación/inmunología , Neoplasias de la Mama/patología , Antígeno CD47/inmunología , Línea Celular Tumoral , Receptores ErbB/antagonistas & inhibidores , Femenino , Humanos , Inmunoterapia/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Receptor ErbB-2/antagonistas & inhibidores , Receptores Inmunológicos/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Front Immunol ; 9: 3124, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30761158

RESUMEN

The function of the low-affinity IgG-receptor FcγRIIIb (CD16b), which is uniquely and abundantly expressed on human granulocytes, is not clear. Unlike the other Fcγ receptors (FcγR), it is a glycophosphatidyl inositol (GPI) -anchored molecule and does not have intracellular signaling motifs. Nevertheless, FcγRIIIb can cooperate with other FcγR to promote phagocytosis of antibody-opsonized microbes by human neutrophils. Here we have investigated the role of FcγRIIIb during antibody-dependent cellular cytotoxicity (ADCC) by neutrophils toward solid cancer cells coated with either trastuzumab (anti-HER2) or cetuximab (anti-EGFR). Inhibiting FcγRIIIb using CD16-F(ab')2 blocking antibodies resulted in substantially enhanced ADCC. ADCC was completely dependent on FcγRIIa (CD32a) and the enhanced ADCC seen after FcγRIIIb blockade therefore suggested that FcγRIIIb was competing with FcγRIIa for IgG on the opsonized target cells. Interestingly, the function of neutrophil FcγRIIIb as a decoy receptor was further supported by using neutrophils from individuals with different gene copy numbers of FCGR3B causing different levels of surface FcγRIIIb expression. Individuals with one copy of FCGR3B showed higher levels of ADCC compared to those with two or more copies. Finally, we show that therapeutic antibodies intended to improve FcγRIIIa (CD16a)-dependent natural killer (NK) cell ADCC due to the lack of fucosylation on the N-linked glycan at position N297 of the IgG1 heavy chain Fc-region, show decreased ADCC as compared to regularly fucosylated antibodies. Together, these data confirm FcγRIIIb as a negative regulator of neutrophil ADCC toward tumor cells and a potential target for enhancing tumor cell destruction by neutrophils.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Inmunoglobulina G/metabolismo , Neoplasias/tratamiento farmacológico , Neutrófilos/inmunología , Receptores de IgG/metabolismo , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Línea Celular Tumoral , Cetuximab/metabolismo , Cetuximab/farmacología , Cetuximab/uso terapéutico , Variaciones en el Número de Copia de ADN , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , Humanos , Inmunoglobulina G/farmacología , Inmunoglobulina G/uso terapéutico , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Neoplasias/patología , Neutrófilos/metabolismo , Receptores de IgG/antagonistas & inhibidores , Receptores de IgG/genética , Receptores de IgG/inmunología , Trastuzumab/metabolismo , Trastuzumab/farmacología , Trastuzumab/uso terapéutico
8.
Cell Rep ; 23(13): 3946-3959.e6, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29949776

RESUMEN

Destruction of cancer cells by therapeutic antibodies occurs, at least in part, through antibody-dependent cellular cytotoxicity (ADCC), and this can be mediated by various Fc-receptor-expressing immune cells, including neutrophils. However, the mechanism(s) by which neutrophils kill antibody-opsonized cancer cells has not been established. Here, we demonstrate that neutrophils can exert a mode of destruction of cancer cells, which involves antibody-mediated trogocytosis by neutrophils. Intimately associated with this is an active mechanical disruption of the cancer cell plasma membrane, leading to a lytic (i.e., necrotic) type of cancer cell death. Furthermore, this mode of destruction of antibody-opsonized cancer cells by neutrophils is potentiated by CD47-SIRPα checkpoint blockade. Collectively, these findings show that neutrophil ADCC toward cancer cells occurs by a mechanism of cytotoxicity called trogoptosis, which can be further improved by targeting CD47-SIRPα interactions.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos , Neutrófilos/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Antígeno CD11b/metabolismo , Antígenos CD18/metabolismo , Antígeno CD47/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/patología , Receptores de IgG/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Trasplante Homólogo
9.
Cell Rep ; 10(10): 1778-1791, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25772364

RESUMEN

Small ubiquitin-like modifiers play critical roles in the DNA damage response (DDR). To increase our understanding of SUMOylation in the mammalian DDR, we employed a quantitative proteomics approach in order to identify dynamically regulated SUMO-2 conjugates and modification sites upon treatment with the DNA damaging agent methyl methanesulfonate (MMS). We have uncovered a dynamic set of 20 upregulated and 33 downregulated SUMO-2 conjugates, and 755 SUMO-2 sites, of which 362 were dynamic in response to MMS. In contrast to yeast, where a response is centered on homologous recombination, we identified dynamically SUMOylated interaction networks of chromatin modifiers, transcription factors, DNA repair factors, and nuclear body components. SUMOylated chromatin modifiers include JARID1B/KDM5B, JARID1C/KDM5C, p300, CBP, PARP1, SetDB1, and MBD1. Whereas SUMOylated JARID1B was ubiquitylated by the SUMO-targeted ubiquitin ligase RNF4 and degraded by the proteasome in response to DNA damage, JARID1C was SUMOylated and recruited to the chromatin to demethylate histone H3K4.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA