Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
Cell Chem Biol ; 31(5): 944-954.e5, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38653243

RESUMO

Agonist antibodies are being pursued for therapeutic applications ranging from neurodegenerative diseases to cancer. For the tumor necrosis factor (TNF) receptor superfamily, higher-order clustering of three or more receptors is key to their activation, which can be achieved using antibodies that recognize two unique epitopes. However, the generation of biepitopic (i.e., biparatopic) antibodies typically requires animal immunization and is laborious and unpredictable. Here, we report a simple method for identifying biepitopic antibodies that potently activate TNF receptors without the need for additional animal immunization. Our approach uses existing, receptor-specific IgGs, which lack intrinsic agonist activity, to block their corresponding epitopes, then selects single-chain antibodies that bind accessible epitopes. The selected antibodies are fused to the light chains of IgGs to generate human tetravalent antibodies. We highlight the broad utility of this approach by converting several clinical-stage antibodies against OX40 and CD137 (4-1BB) into biepitopic antibodies with potent agonist activity.


Assuntos
Epitopos , Humanos , Epitopos/imunologia , Epitopos/química , Animais , Receptores do Fator de Necrose Tumoral/agonistas , Receptores do Fator de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/metabolismo , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/agonistas , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/antagonistas & inibidores , Receptores OX40/agonistas , Receptores OX40/imunologia , Receptores OX40/metabolismo , Receptores OX40/antagonistas & inibidores , Anticorpos/imunologia , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/farmacologia , Camundongos
3.
FEBS Open Bio ; 12(8): 1542-1557, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35674216

RESUMO

Glucocorticoid-induced TNF receptor-related (GITR) can act as a co-stimulatory receptor, representing a potential target for safely enhancing immunotherapy efficacy. GITR is triggered by a GITR ligand or an agonist antibody and activates CD8+ and CD4+ effector T cells, reducing tumor-infiltrating Treg numbers and resulting in activation of immune responses and tumor cell destruction by effector T cells. GITR is an attractive target for immunotherapy, especially in combination therapy with immune checkpoint inhibitors, as is being explored in clinical trials. Using H2L2 transgenic mice encoding the human immunoglobulin variable region and hybridoma technology, we generated a panel of fully human antibodies that showed excellent specific affinity and strong activation of human T cells. After conversion to fully human antibodies and engineering modification, we obtained an anti-GITR antibody hab019e2 with enhanced antitumor activity in a B-hGITR MC38 mouse model compared to Tab9H6V3, an anti-GITR antibody that activates T cells and inhibits Treg suppression from XenoMouse. As a fully human antibody with its posttranslational modification hot spot removed, the hab019e2 antibody exerted more potent therapeutic effects, and may have potential as a novel and developable antibody targeting GITR for follow-up drug studies.


Assuntos
Glucocorticoides , Receptores do Fator de Necrose Tumoral , Animais , Anticorpos , Linhagem Celular Tumoral , Proteína Relacionada a TNFR Induzida por Glucocorticoide/agonistas , Humanos , Imunoterapia/métodos , Camundongos , Receptores do Fator de Necrose Tumoral/agonistas
4.
Cell Rep Med ; 3(6): 100660, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35732156

RESUMO

Glucocorticoid-induced tumor necrosis factor receptor (GITR) is a co-stimulatory receptor and an important target for cancer immunotherapy. We herein present a potent FcγR-independent GITR agonist IBI37G5 that can effectively activate effector T cells and synergize with anti-programmed death 1 (PD1) antibody to eradicate established tumors. IBI37G5 depends on both antibody bivalency and GITR homo-dimerization for efficient receptor cross-linking. Functional analyses reveal bell-shaped dose responses due to the unique 2:2 antibody-receptor stoichiometry required for GITR activation. Antibody self-competition is observed after concentration exceeded that of 100% receptor occupancy (RO), which leads to antibody monovalent binding and loss of activity. Retrospective pharmacokinetics/pharmacodynamics analysis demonstrates that the maximal efficacy is achieved at medium doses with drug exposure near saturating GITR occupancy during the dosing cycle. Finally, we propose an alternative dose-finding strategy that does not rely on the traditional maximal tolerated dose (MTD)-based paradigm but instead on utilizing the RO-function relations as biomarker to guide the clinical translation of GITR and similar co-stimulatory agonists.


Assuntos
Glucocorticoides , Receptores de IgG , Linhagem Celular Tumoral , Proteína Relacionada a TNFR Induzida por Glucocorticoide/agonistas , Ligantes , Receptores do Fator de Necrose Tumoral/agonistas , Estudos Retrospectivos , Fatores de Necrose Tumoral
5.
Nat Cancer ; 3(3): 337-354, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35256819

RESUMO

Costimulatory receptors such as glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR) play key roles in regulating the effector functions of T cells. In human clinical trials, however, GITR agonist antibodies have shown limited therapeutic effect, which may be due to suboptimal receptor clustering-mediated signaling. To overcome this potential limitation, a rational protein engineering approach is needed to optimize GITR agonist-based immunotherapies. Here we show a bispecific molecule consisting of an anti-PD-1 antibody fused with a multimeric GITR ligand (GITR-L) that induces PD-1-dependent and FcγR-independent GITR clustering, resulting in enhanced activation, proliferation and memory differentiation of primed antigen-specific GITR+PD-1+ T cells. The anti-PD-1-GITR-L bispecific is a PD-1-directed GITR-L construct that demonstrated dose-dependent, immunologically driven tumor growth inhibition in syngeneic, genetically engineered and xenograft humanized mouse tumor models, with a dose-dependent correlation between target saturation and Ki67 and TIGIT upregulation on memory T cells. Anti-PD-1-GITR-L thus represents a bispecific approach to directing GITR agonism for cancer immunotherapy.


Assuntos
Neoplasias , Receptor de Morte Celular Programada 1 , Animais , Análise por Conglomerados , Modelos Animais de Doenças , Proteína Relacionada a TNFR Induzida por Glucocorticoide/agonistas , Humanos , Imunoterapia/métodos , Camundongos , Neoplasias/tratamento farmacológico , Receptores do Fator de Necrose Tumoral/agonistas , Linfócitos T
6.
Commun Biol ; 4(1): 772, 2021 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-34162985

RESUMO

Monoclonal antibodies (mAb) and natural ligands targeting costimulatory tumor necrosis factor receptors (TNFR) exhibit a wide range of agonistic activities and antitumor responses. The mechanisms underlying these differential agonistic activities remain poorly understood. Here, we employ a panel of experimental and clinically-relevant molecules targeting human CD40, 4-1BB and OX40 to examine this issue. Confocal and STORM microscopy reveal that strongly agonistic reagents induce clusters characterized by small area and high receptor density. Using antibody pairs differing only in isotype we show that hIgG2 confers significantly more receptor clustering than hIgG1 across all three receptors, explaining its greater agonistic activity, with receptor clustering shielding the receptor-agonist complex from further molecular access. Nevertheless, discrete receptor clustering patterns are observed with different hIgG2 mAb, with a unique rod-shaped assembly observed with the most agonistic mAb. These findings dispel the notion that larger receptor clusters elicit greater agonism, and instead point to receptor density and subsequent super-structure as key determinants.


Assuntos
Receptores do Fator de Necrose Tumoral/agonistas , Animais , Anticorpos Monoclonais/farmacologia , Afinidade de Anticorpos , Antígenos CD40/agonistas , Antígenos CD40/química , Linhagem Celular , Humanos , Imunoglobulina G/farmacologia , Camundongos , Microscopia Confocal , Receptores OX40/agonistas , Receptores do Fator de Necrose Tumoral/química , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/agonistas
7.
J Immunother Cancer ; 7(1): 191, 2019 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-31324216

RESUMO

BACKGROUND: Glucocorticoid-induced TNFR-related protein (TNFRSF18, GITR, CD357), expressed by T cells, and its ligand (TNFSF18, GITRL), expressed by myeloid populations, provide co-stimulatory signals that boost T cell activity. Due to the important role that GITR plays in regulating immune functions, agonistic stimulation of GITR is a promising therapeutic concept. Multiple strategies to induce GITR signaling have been investigated. The limited clinical efficacy of antibody-based GITR agonists results from structural and functional characteristics of antibodies that are unsuitable for stimulating the well-defined trimeric members of the TNFRSF. METHODS: To overcome limitations of antibody-based TNFRSF agonists, we have developed HERA-GITRL, a fully human hexavalent TNF receptor agonist (HERA) targeting GITR and mimicking the natural signaling concept. HERA-GITRL is composed of a trivalent but single-chain GITRL-receptor-binding-domain (scGITRL-RBD) unit fused to an IgG1 derived silenced Fc-domain serving as dimerization scaffold. A specific mouse surrogate, mmHERA-GITRL, was also generated to examine in vivo activity in respective mouse tumor models. RESULTS: For functional characterization of HERA-GITRL in vitro, human immune cells were isolated from healthy-donor blood and stimulated with anti-CD3 antibody in the presence of HERA-GITRL. Consistently, HERA-GITRL increased the activity of T cells, including proliferation and differentiation, even in the presence of regulatory T cells. In line with these findings, mmHERA-GITRL enhanced antigen-specific clonal expansion of both CD4+ (OT-II) and CD8+ (OT-I) T cells in vivo while having no effect on non-specific T cells. In addition, mmHERA-GITRL showed single-agent anti-tumor activity in two subcutaneous syngeneic colon cancer models (CT26wt and MC38-CEA). Importantly, this activity is independent of its FcγR-binding functionality, as both mmHERA-GITRL with a functional Fc- and a silenced Fc-domain showed similar tumor growth inhibition. Finally, in a direct in vitro comparison to a bivalent clinical benchmark anti-GITR antibody and a trivalent GITRL, only the hexavalent HERA-GITRL showed full biological activity independent of additional crosslinking. CONCLUSION: In this manuscript, we describe the development of HERA-GITRL, a true GITR agonist with a clearly defined mechanism of action. By clustering six receptor chains in a spatially well-defined manner, HERA-GITRL induces potent agonistic activity without being dependent on additional FcγR-mediated crosslinking.


Assuntos
Receptores do Fator de Necrose Tumoral/agonistas , Anticorpos de Cadeia Única/administração & dosagem , Linfócitos T Reguladores/imunologia , Fatores de Necrose Tumoral/química , Animais , Linhagem Celular Tumoral , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Ativação Linfocitária , Macaca fascicularis , Camundongos , Proteínas Recombinantes de Fusão/imunologia , Transdução de Sinais , Anticorpos de Cadeia Única/imunologia , Fatores de Necrose Tumoral/metabolismo
8.
BioDrugs ; 32(3): 221-231, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29637478

RESUMO

Recent advancements in T-cell biology and antibody engineering have opened doors to significant improvements in cancer immunotherapy. Initial success with monoclonal antibodies targeting key receptors that inhibit T-cell function such as cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed death-ligand 1 (PD-1) have demonstrated the potency of this new class of therapy, highlighted by long-term complete responses for metastatic cancers once thought incurable. However, only a subset of patients responds to checkpoint blockade because of a multitude of factors, including an immunosuppressive tumor microenvironment and the mutational burden of the cancer. Novel antibodies, as well as ligand-immunoglobulin fusion proteins that target costimulatory immune receptors, are being developed and tested in clinical trials to further enhance the anti-tumor immune response. Many of these costimulatory receptors are in the tumor necrosis factor receptor superfamily (TNFRSF) and are expressed on multiple immune cell types, including inhibitory cells. While TNFRSFs signal through common pathways, the outcome of targeting different receptors depends on the functional status of the cell types expressing the relevant receptors. In this review, we discuss the current state of targeted costimulatory immunotherapy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Receptores Coestimuladores e Inibidores de Linfócitos T/imunologia , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Linfócitos T/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Antígeno CTLA-4/antagonistas & inibidores , Antígeno CTLA-4/imunologia , Ensaios Clínicos como Assunto , Humanos , Ativação Linfocitária , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Receptores do Fator de Necrose Tumoral/agonistas , Receptores do Fator de Necrose Tumoral/imunologia
9.
Sci Rep ; 7(1): 13301, 2017 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-29038485

RESUMO

TNFR superfamily (TNFRSF) members have important immunoregulatory functions and are of clear interest for cancer immunotherapy. Various TNFRSF agonists have been clinically evaluated, but have met with limited efficacy and/or toxicity. Recent insights indicate that 'first-generation' TNFRSF agonists lack efficacy as they do not effectively cross-link their corresponding receptor. Reversely, ubiquitous TNFRSF receptor(s) cross-linking by CD40 and Fas agonistic antibodies resulted in dose-limiting liver toxicity. To overcome these issues, we developed a novel pretargeting strategy exploiting recombinant fusion proteins in which a soluble form of TRAIL, FasL or CD40L is genetically fused to a high-affinity anti-fluorescein scFv antibody fragment (scFvFITC). Fusion proteins scFvFITC:sTRAIL and scFvFITC:sFasL induced potent target antigen-restricted apoptosis in a panel of cancer lines and in primary patient-derived cancer cells, but only when pretargeted with a relevant FITC-labelled antitumour antibody. In a similar pretargeting setting, fusion protein scFvFITC:sCD40L promoted tumour-directed maturation of immature monocyte-derived dendritic cells (iDCs). This novel tumour-selective pretargeting approach may be used to improve efficacy and/or reduce possible off-target toxicity of TNFSF ligands for cancer immunotherapy.


Assuntos
Anticorpos Monoclonais/farmacologia , Receptores do Fator de Necrose Tumoral/agonistas , Anticorpos de Cadeia Única/farmacologia , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/química , Apoptose/efeitos dos fármacos , Biomarcadores , Ligante de CD40/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citotoxicidade Imunológica , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Proteína Ligante Fas/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Leucemia , Modelos Biológicos , Terapia de Alvo Molecular , Ligação Proteica , Anticorpos de Cadeia Única/administração & dosagem , Anticorpos de Cadeia Única/química , Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores
10.
Curr Opin Immunol ; 47: 70-77, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28750279

RESUMO

Cancer vaccines have offered unrequited hope as a mechanism for rapidly and potently eliciting a patient's immune system to counter tumors. Initial results from preclinical mouse models have not translated to substantial benefit to patients, suggesting that either the targets or the vaccination approach were inadequate. Recent innovations in antigen identification have spiked renewed interest vaccination technologies. This has coincided with a detailed molecular understanding of the coordinated steps in postactivation support of T cell proliferation, differentiation and survival, leading to the development of novel targets and combinations that are substantially more effective than first and second generation cancer vaccines in preclinical models. Within this cluster of developments, the TNF-receptor superfamily members have emerged as attractive candidates for clinical implementation. Here we review recent developments in the mechanisms of action of TNFRSF agonists, and how their activity is potentiated by integration co-targeting pattern recognition receptors.


Assuntos
Adjuvantes Imunológicos/farmacologia , Vacinas Anticâncer/imunologia , Receptores do Fator de Necrose Tumoral/agonistas , Vacinação , Adjuvantes Imunológicos/uso terapêutico , Animais , Modelos Animais de Doenças , Humanos , Camundongos
11.
Hum Antibodies ; 25(3-4): 87-109, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28085016

RESUMO

Co-stimulatory tumor necrosis factor receptors (TNFRs) can sculpt the responsiveness of T cells recognizing tumor-associated antigens. For this reason, agonist antibodies targeting CD137, CD357, CD134 and CD27 have received considerable attention for their therapeutic utility in enhancing anti-tumor immune responses, particularly in combination with other immuno-modulatory antibodies targeting co-inhibitory pathways in T cells. The design of therapeutic antibodies that optimally engage and activate co-stimulatory TNFRs presents an important challenge of how to promote effective anti-tumor immunity while avoiding serious immune-related adverse events. Here we review our current understanding of the expression, signaling and structural features of CD137, CD357, CD134 and CD27, and how this may inform the design of pharmacologically active immuno-modulatory antibodies targeting these receptors. This includes the integration of our emerging knowledge of the role of Fcγ receptors (FcγRs) in facilitating antibody-mediated receptor clustering and forward signaling, as well as promoting immune effector cell-mediated activities. Finally, we bring our current preclinical and clinical knowledge of co-stimulatory TNFR antibodies into the context of opportunities for next generation molecules with improved pharmacologic properties.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Receptores de IgG/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Regulação da Expressão Gênica , Humanos , Imunidade Celular/efeitos dos fármacos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Receptores de IgG/agonistas , Receptores de IgG/genética , Receptores do Fator de Necrose Tumoral/agonistas , Receptores do Fator de Necrose Tumoral/genética , Transdução de Sinais , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/patologia
12.
J Control Release ; 234: 104-14, 2016 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-27212104

RESUMO

Receptor clustering is important for signaling among the therapeutically relevant TNFR superfamily of receptors. In nature, this clustering is driven by trimeric ligands often presented in large numbers as cell surface proteins. Molecules capable of driving similar levels of clustering could make good agonists and hold therapeutic value. However, recapitulating such extensive clustering using typical biotherapeutic formats, such as antibodies, is difficult. Consequently, generating effective agonists of TNFR superfamily receptors is challenging. Toward addressing this challenge we have used lipid- and polyion complex-based micelles as platforms for presenting receptor-binding biologics in a multivalent format that facilitates receptor clustering and imparts strong agonist activity. We show that receptor-binding scFvs or small antibody mimetics that have no agonist activity on their own can be transformed into potent agonists through multivalent presentation on a micelle surface and that the activity of already active multivalent agonists can be enhanced. Using this strategy, we generated potent agonists against two different TNFR superfamily receptors and mouse tumor model studies demonstrate that these micellar agonists have therapeutic efficacy in vivo. Due to its ease of implementation and applicability independent of agonist molecular format, we anticipate that this strategy could be useful for developing agonists to a variety of receptors that rely on clustering to signal.


Assuntos
Antineoplásicos/administração & dosagem , Maleimidas/química , Nanopartículas/química , Fosfatidiletanolaminas/química , Polietilenoglicóis/química , Receptores do Fator de Necrose Tumoral/agonistas , Anticorpos de Cadeia Única/administração & dosagem , Animais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Humanos , Células Jurkat , Camundongos , Micelas , Ligação Proteica , Anticorpos de Cadeia Única/química , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Methods Mol Biol ; 1280: 103-19, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25736746

RESUMO

The alternative or noncanonical NF-κB pathway regulates the generation of p52-containing NF-κB dimers (e.g., p52/RelB) through a partial degradation (called processing) of the precursor p100 into p52. This pathway is activated by a subset of non-death TNF receptor members, which ultimately activate two kinases: NIK (NF-κB-Inducing Kinase) and IKKα (Inhibitor of κB Kinase alpha). These kinases create a phosphodegron for the E3 ligase SCF-ß-TrCP that covalently binds K48-linked polyubiquitin chain onto p100 prior to its proteasomal processing. The resulting p52-containing complexes translocate into the nucleus to activate target genes involved in secondary lymphoid organ development, B cell survival or in osteoclastogenesis. We describe in this chapter straightforward methods to monitor the activation of the alternative NF-κB pathway. These methods uncover cytosolic and nuclear biochemical modifications of key proteins of the alternative NF-κB pathway required prior to the transcription of NF-κB target genes.


Assuntos
NF-kappa B/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Anticorpos Monoclonais/farmacologia , Ensaio de Desvio de Mobilidade Eletroforética , Ativação Enzimática , Estabilidade Enzimática , Humanos , Quinase I-kappa B/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Ligantes , Subunidade p52 de NF-kappa B/metabolismo , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , Proteólise , Receptores do Fator de Necrose Tumoral/agonistas , Transdução de Sinais/efeitos dos fármacos , Fator 2 Associado a Receptor de TNF/metabolismo , Fator 3 Associado a Receptor de TNF/metabolismo , Fator de Transcrição RelB/metabolismo , Quinase Induzida por NF-kappaB
14.
Methods Mol Biol ; 1280: 269-82, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25736754

RESUMO

Precise regulation of survival and signaling pathways is essential for proper maintenance of organismal homeostasis, development, and immune defense. Inhibitor of apoptosis (IAP) proteins are evolutionarily conserved regulators of cell death and immune signaling that impact numerous cellular processes. Initially characterized as inhibitors of apoptosis, the ubiquitin ligase activity of IAP proteins is critical for modulating various signaling pathways (e.g., NF-κB, MAPK) and cellular fate. Cellular IAP1 and IAP2 regulate the pro-survival canonical NF-κB pathway by ubiquitinating RIP1 and enabling recruitment of kinase (IKK) and E3 ligase (LUBAC) complexes. On the other hand, c-IAP1 and c-IAP2 are negative regulators of noncanonical NF-κB signaling by promoting ubiquitination and consequent degradation of the NF-κB-inducing kinase NIK. In this article, we describe the involvement of c-IAP1 and c-IAP2 in NF-κB signaling and provide detailed methodology for examining how c-IAPs exert their functional roles.


Assuntos
Proteínas Inibidoras de Apoptose/metabolismo , NF-kappa B/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Western Blotting/métodos , Linhagem Celular , Ativação Enzimática , Expressão Gênica , Humanos , Imunoprecipitação/métodos , Ligantes , Proteínas Quinases Ativadas por Mitógeno/metabolismo , RNA Interferente Pequeno/genética , Receptores do Fator de Necrose Tumoral/agonistas , Frações Subcelulares , Transfecção
15.
Semin Immunol ; 26(3): 210-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24910294

RESUMO

Members of the TNFR family can play prominent roles in controlling the magnitude, duration and phenotype of the immune response to viruses. The importance of particular TNFRs in different viral infections and whether they contribute to viral control or pathology is dependent on the virus and the severity of the infection. TNFRs and their ligands are widely and differentially expressed on both adaptive and innate immune cell types. The cell types through which TNFRs exert their effects, the unique signals provided by each member of the family, and how these signals are ultimately integrated during an anti-viral immune response remain to be fully elucidated. Here we discuss the role of 4-1BB, OX40, CD27 and GITR and their ligands during viral infection and highlight some of the outstanding questions in the field.


Assuntos
Receptores do Fator de Necrose Tumoral/metabolismo , Linfócitos T/imunologia , Viroses/imunologia , Animais , Humanos , Receptores do Fator de Necrose Tumoral/agonistas , Linfócitos T/metabolismo , Viroses/tratamento farmacológico , Viroses/patologia , Viroses/virologia
16.
Cancer Immunol Immunother ; 62(5): 941-8, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23543215

RESUMO

Isotype plays a crucial role in therapeutic monoclonal antibody (mAb) function, mediated in large part through differences in Fcγ receptor (FcγR) interaction. Monoclonal Abs such as rituximab and alemtuzumab, which bind target cells directly, are designed for efficient recruitment of immune effector cells through their activatory FcγR engagement to mediate maximal target cell killing. In this setting, binding to inhibitory FcγRIIB is thought to inhibit function, making mAbs with high activatory/inhibitory (A/I) FcγR binding ratios, such as mouse IgG2a and human IgG1, the first choice for this role. In contrast, exciting new data show that agonistic mAbs directed against the tumour necrosis factor receptor superfamily member CD40 require interaction with FcγRIIB for in vivo function. Such ligation activates antigen-presenting cells, promotes myeloid and CTL responses and potentially stimulates effective anti-cancer immunity. It appears that the role of FcγRIIB is to mediate mAb hyper-crosslinking to allow CD40 downstream intracellular signalling. Previous work has shown that mAbs directed against other TNFR family members, Fas and death receptor 5 and probably death receptor 4, also require FcγRIIB hyper-crosslinking to promote target cell apoptosis, suggesting a common mechanism of action. In mouse models, IgG1 is optimal for these agents as it binds to FcγRIIB with tenfold higher affinity than IgG2a and hence has a relatively low A:I FcγR binding ratio. In contrast, human IgG isotypes have a universally low affinity for FcγRIIB, but in the case of human IgG1, engineering the Fc to increase its affinity for FcγRIIB can potentially overcome this problem. Thus, modifying the A/I binding ratio of human IgG Fc can be used to optimise different types of therapeutic activity by enhancing cytotoxic or hyper-crosslinking function.


Assuntos
Anticorpos Monoclonais/imunologia , Neoplasias/terapia , Receptores de IgG/imunologia , Receptores do Fator de Necrose Tumoral/agonistas , Receptores do Fator de Necrose Tumoral/imunologia , Animais , Apoptose , Antígenos CD40/metabolismo , Humanos , Imunoglobulina G/imunologia , Camundongos , Neoplasias/imunologia , Ligação Proteica , Transdução de Sinais
17.
Clin Cancer Res ; 19(5): 1044-53, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23460535

RESUMO

Therapy for cancer can be achieved by artificially stimulating antitumor T and natural killer (NK) lymphocytes with agonist monoclonal antibodies (mAb). T and NK cells express several members of the TNF receptor (TNFR) family specialized in delivering a costimulatory signal on their surface. Engagement of these receptors is typically associated with proliferation, elevated effector functions, resistance to apoptosis, and differentiation into memory cells. These receptors lack any intrinsic enzymatic activity and their signal transduction relies on associations with TNFR-associated factor (TRAF) adaptor proteins. Stimulation of CD137 (4-1BB), CD134 (OX40), and glucocorticoid-induced TNFR (GITR; CD357) promotes impressive tumor-rejecting immunity in a variety of murine tumor models. The mechanisms of action depend on a complex interplay of CTL, T-helper cells, regulatory T cells, dendritic cells, and vascular endothelium in tumors. Agonist mAbs specific for CD137 have shown signs of objective clinical activity in patients with metastatic melanoma, whereas anti-OX40 and anti-GITR mAbs have entered clinical trials. Preclinical evidence suggests that engaging TNFR members would be particularly active with conventional cancer therapies and additional immunotherapeutic approaches. Indeed, T-cell responses elicited to tumor antigens by means of immunogenic tumor cell death are amplified by these immunostimulatory agonist mAbs. Furthermore, anti-CD137 mAbs have been shown to enhance NK-mediated cytotoxicity elicited by rituximab and trastuzumab. Combinations with other immunomodulatory mAb that block T-cell checkpoint blockade receptors such as CTLA-4 and PD-1 are also promising.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Imunoterapia , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Neoplasias/terapia , Receptores do Fator de Necrose Tumoral/agonistas , Animais , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Neoplasias/imunologia , Neoplasias/metabolismo , Receptores do Fator de Necrose Tumoral/imunologia
18.
Gynecol Oncol ; 127(2): 426-32, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22885470

RESUMO

Surgical treatment including total abdominal hysterectomy+bilateral salpingo oopherectomy (TAH+BSO) with pelvic and para-aortic lymphadenectomy may not be sufficient to treat cases with advanced endometrial adenocarcinoma (EAC), and in these cases, adjuvant treatments including radiotherapy and/or chemotherapy, are employed based upon the tumor location, type and stage of the disease. These treatment modalities have high incidence of systemic toxicity, thereby compelling clinicians to look for targeted therapy aiming specifically at the malignant cells. Bevacizumab (anti-VEGF), temsirolimus (mTOR inhibitor) and aflibercept (VEGF trap) are already under clinical trials in women with EAC. Targeting the ligands and receptors of the tumor necrosis factor (TNF) superfamily holds promise in this regard. The objective of this review is to provide an overview of the various mechanisms and pathways related to the TNF superfamily involved in advanced EAC and to identify the new therapeutic strategies for specifically targeting these impaired pathways. In addition, the development of treatments for EAC is also discussed. The possible therapeutic treatments include targeting TNFα and its receptors using monoclonal antibodies (MAbs) such as infliximab, adalimumab, etanercept, and certolizumab. Proteosome inhibitors including bortezomib and the anti CD-20 agent rituximab are used to inhibit the NF-κB pathway. Other options include targeting the FAS (CD95) pathway and the TNF-related apoptosis-inducing ligand (TRAIL) pathway using agents such as mapatumab, lexatumumab, and conatumumab. These pathways are known to be involved in the pathogenesis of EAC. Moreover, there is adequate evidence to warrant the use of drugs that target the TNF superfamily for the treatment of advanced EAC.


Assuntos
Adenocarcinoma/tratamento farmacológico , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias do Endométrio/tratamento farmacológico , Terapia de Alvo Molecular , Receptores do Fator de Necrose Tumoral , Fatores de Necrose Tumoral , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma/cirurgia , Biomarcadores Tumorais/metabolismo , Quimioterapia Adjuvante , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Neoplasias do Endométrio/cirurgia , Feminino , Humanos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Estadiamento de Neoplasias , Receptores do Fator de Necrose Tumoral/agonistas , Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Receptores do Fator de Necrose Tumoral/metabolismo , Inibidores do Fator de Necrose Tumoral , Fatores de Necrose Tumoral/agonistas , Fatores de Necrose Tumoral/metabolismo
20.
MAbs ; 3(4): 362-75, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21697654

RESUMO

TWEAK, a TNF family ligand with pleiotropic cellular functions, was originally described as capable of inducing tumor cell death in vitro. TWEAK functions by binding its receptor, Fn14, which is up-regulated on many human solid tumors. Herein, we show that intratumoral administration of TWEAK, delivered either by an adenoviral vector or in an immunoglobulin Fc-fusion form, results in significant inhibition of tumor growth in a breast xenograft model. To exploit the TWEAK-Fn14 pathway as a therapeutic target in oncology, we developed an anti-Fn14 agonistic antibody, BIIB036. Studies described herein show that BIIB036 binds specifically to Fn14 but not other members of the TNF receptor family, induces Fn14 signaling, and promotes tumor cell apoptosis in vitro. In vivo, BIIB036 effectively inhibits growth of tumors in multiple xenograft models, including colon (WiDr), breast (MDA-MB-231), and gastric (NCI-N87) tumors, regardless of tumor cell growth inhibition response observed to BIIB036 in vitro. The anti-tumor activity in these cell lines is not TNF-dependent. Increasing the antigen-binding valency of BIB036 significantly enhances its anti-tumor effect, suggesting the contribution of higher order cross-linking of the Fn14 receptor. Full Fc effector function is required for maximal activity of BIIB036 in vivo, likely due to the cross-linking effect and/or ADCC mediated tumor killing activity. Taken together, the anti-tumor properties of BIIB036 validate Fn14 as a promising target in oncology and demonstrate its potential therapeutic utility in multiple solid tumor indications.


Assuntos
Antineoplásicos , Apoptose/efeitos dos fármacos , Neoplasias/terapia , Receptores do Fator de Necrose Tumoral/agonistas , Fatores de Necrose Tumoral/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacologia , Antineoplásicos/administração & dosagem , Antineoplásicos/imunologia , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Citocina TWEAK , Células HT29 , Humanos , Ligantes , Camundongos , Neoplasias/imunologia , Ligação Proteica , Receptores do Fator de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/metabolismo , Receptor de TWEAK , Fatores de Necrose Tumoral/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA