Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cancer Immunol Res ; 9(10): 1141-1157, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34376502

RESUMO

The use of cytokines for immunotherapy shows clinical efficacy but is frequently accompanied by severe adverse events caused by excessive and systemic immune activation. Here, we set out to address these challenges by engineering a fusion protein of a single, potency-reduced, IL15 mutein and a PD1-specific antibody (anti-PD1-IL15m). This immunocytokine was designed to deliver PD1-mediated, avidity-driven IL2/15 receptor stimulation to PD1+ tumor-infiltrating lymphocytes (TIL) while minimally affecting circulating peripheral natural killer (NK) cells and T cells. Treatment of tumor-bearing mice with a mouse cross-reactive fusion, anti-mPD1-IL15m, demonstrated potent antitumor efficacy without exacerbating body weight loss in B16 and MC38 syngeneic tumor models. Moreover, anti-mPD1-IL15m was more efficacious than an IL15 superagonist, an anti-mPD-1, or the combination thereof in the B16 melanoma model. Mechanistically, anti-PD1-IL15m preferentially targeted CD8+ TILs and single-cell RNA-sequencing analyses revealed that anti-mPD1-IL15m treatment induced the expansion of an exhausted CD8+ TIL cluster with high proliferative capacity and effector-like signatures. Antitumor efficacy of anti-mPD1-IL15m was dependent on CD8+ T cells, as depletion of CD8+ cells resulted in the loss of antitumor activity, whereas depletion of NK cells had little impact on efficacy. The impact of anti-hPD1-IL15m on primary human TILs from patients with cancer was also evaluated. Anti-hPD1-IL15m robustly enhanced the proliferation, activation, and cytotoxicity of CD8+ and CD4+ TILs from human primary cancers in vitro, whereas tumor-derived regulatory T cells were largely unaffected. Taken together, our findings showed that anti-PD1-IL15m exhibits a high translational promise with improved efficacy and safety of IL15 for cancer immunotherapy via targeting PD1+ TILs.See related Spotlight by Felices and Miller, p. 1110.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Neoplasias do Colo/terapia , Imunoterapia , Interleucina-15/uso terapêutico , Melanoma Experimental/terapia , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Modelos Animais de Doenças , Humanos , Interleucina-15/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/imunologia , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico
2.
J Exp Med ; 218(8)2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34106206

RESUMO

As indicated by its name, V-domain Ig suppressor of T cell activation (VISTA) is thought to serve primarily as an inhibitory protein that limits immune responses. VISTA antibodies can dampen the effects of several concomitantly elicited activation signals, including TCR and TLR activation, but it is currently unclear if VISTA agonism could singly affect immune cell biology. In this study, we discovered two novel VISTA antibodies and characterized their effects on human peripheral blood mononuclear cells by scRNA/CITE-seq. Both antibodies appeared to agonize VISTA in an Fc-functional manner to elicit transcriptional and functional changes in monocytes consistent with activation. We also used pentameric VISTA to identify Syndecan-2 and several heparan sulfate proteoglycan synthesis genes as novel regulators of VISTA interactions with monocytic cells, adding further evidence of bidirectional signaling. Together, our study highlights several novel aspects of VISTA biology that have yet to be uncovered in myeloid cells and serves as a foundation for future research.


Assuntos
Antígenos B7/metabolismo , Monócitos/metabolismo , Receptores Imunológicos/metabolismo , Anticorpos Monoclonais/metabolismo , Especificidade de Anticorpos/imunologia , Sistemas CRISPR-Cas/genética , Heparitina Sulfato/metabolismo , Humanos , Ligação Proteica , Receptores Fc/metabolismo , Sindecana-2/metabolismo , Transcrição Gênica , Transcriptoma/genética
3.
J Immunol ; 197(5): 1754-61, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27439516

RESUMO

As a result of the growing burden of tumors and chronic infections, manipulating CD8 T cell responses for clinical use has become an important goal for immunologists. In this article, we show that dendritic cell (DC) immunization coupled with relatively early (days 1-3) or late (days 4-6) administration of enhanced IL-2 signals increase peak effector CD8 T cell numbers, but only early IL-2 signals enhance memory numbers. IL-2 signals delivered at relatively late time points drive terminal differentiation and marked Bim-mediated contraction and do not increase memory T cell numbers. In contrast, early IL-2 signals induce effector cell metabolic profiles that are more conducive to memory formation. Of note, downregulation of CD80 and CD86 was observed on DCs in vivo following early IL-2 treatment. Mechanistically, early IL-2 treatment enhanced CTLA-4 expression on regulatory T cells, and CTLA-4 blockade alongside IL-2 treatment in vivo prevented the decrease in CD80 and CD86, supporting a cell-extrinsic role for CTLA-4 in downregulating B7 ligand expression on DCs. Finally, DC immunization followed by early IL-2 treatment and anti-CTLA-4 blockade resulted in lower memory CD8 T cell numbers compared with the DC+early IL-2 treatment group. These data suggest that curtailed signaling through the B7-CD28 costimulatory axis during CD8 T cell activation limits terminal differentiation and preserves memory CD8 T cell formation; thus, it should be considered in future T cell-vaccination strategies.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Memória Imunológica , Interleucina-2/metabolismo , Transdução de Sinais , Animais , Antígenos B7/genética , Antígenos B7/imunologia , Antígeno B7-1/genética , Antígeno B7-1/imunologia , Antígeno B7-2/imunologia , Antígenos CD28/imunologia , Antígenos CD28/metabolismo , Antígeno CTLA-4/genética , Antígeno CTLA-4/imunologia , Contagem de Células , Regulação para Baixo , Imunização , Interleucina-2/imunologia , Ativação Linfocitária , Camundongos , Linfócitos T Reguladores/imunologia , Fatores de Tempo
4.
PLoS Pathog ; 11(10): e1005199, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26431533

RESUMO

Memory CD8 T cells provide protection to immune hosts by eliminating pathogen-infected cells during re-infection. While parameters influencing the generation of primary (1°) CD8 T cells are well established, the factors controlling the development of secondary (2°) CD8 T cell responses remain largely unknown. Here, we address the mechanisms involved in the generation and development of 2° memory (M) CD8 T cells. We observed that the time at which 1° M CD8 T cells enter into immune response impacts their fate and differentiation into 2° M CD8 T cells. Late-entry of 1° M CD8 T cells into an immune response (relative to the onset of infection) not only facilitated the expression of transcription factors associated with memory formation in 2° effector CD8 T cells, but also influenced the ability of 2° M CD8 T cells to localize within the lymph nodes, produce IL-2, and undergo Ag-driven proliferation. The timing of stimulation of 1° M CD8 T cells also impacted the duration of expression of the high-affinity IL-2 receptor (CD25) on 2° effector CD8 T cells and their sensitivity to IL-2 signaling. Importantly, by blocking or enhancing IL-2 signaling in developing 2° CD8 T cells, we provide direct evidence for the role of IL-2 in controlling the differentiation of Ag-driven 2° CD8 T cell responses. Thus, our data suggest that the process of 1° M to 2° M CD8 T cell differentiation is not fixed and can be manipulated, a notion with relevance for the design of future prime-boost vaccination approaches.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Memória Imunológica/imunologia , Interleucina-2/imunologia , Ativação Linfocitária/imunologia , Transdução de Sinais/imunologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/imunologia , Citometria de Fluxo , Immunoblotting , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Fatores de Tempo
5.
Adv Exp Med Biol ; 850: 31-41, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26324344

RESUMO

Antigen-specific CD8 T cells provide an important protective role in response to infection by viruses, intracellular bacteria, and parasites. Pathogen-specific CD8 T cells render this protection by undergoing robust expansion in numbers while gaining the ability to produce cytokines and cytolytic machinery. Creating optimal CD8 T cell responses to infection can be critical for raising sufficient armament to provide protection against invading intracellular pathogens. Although CD8 T cells have protective value, many vaccine strategies tend to focus on creating productive B cell antibody responses to promote immunological protection. Even though antibody responses can be highly protective, coupling optimal CD8 T cell responses with suboptimal B cell responses could provide higher orders of protection than either one on their own. Therefore, a deeper understanding of the pathways that ultimately guide the magnitude of CD8 T cell responses is required to explore this potential therapeutic benefit. The following chapter highlights our current understanding of how inflammatory cytokines regulate the magnitude of CD8 T cell responses.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Doenças Transmissíveis/imunologia , Inflamação/imunologia , Interferon-alfa/imunologia , Interferon beta/imunologia , Interleucina-12/imunologia , Neoplasias/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/patologia , Linfócitos T CD8-Positivos/patologia , Proliferação de Células , Doenças Transmissíveis/genética , Doenças Transmissíveis/metabolismo , Doenças Transmissíveis/patologia , Regulação da Expressão Gênica , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Interferon-alfa/genética , Interferon beta/genética , Interferon gama/genética , Interferon gama/imunologia , Interleucina-12/genética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Transdução de Sinais
6.
J Exp Med ; 211(1): 105-20, 2014 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-24367005

RESUMO

TCR ligation and co-stimulation induce cellular division; however, optimal accumulation of effector CD8 T cells requires direct inflammatory signaling by signal 3 cytokines, such as IL-12 or type I IFNs. Although in vitro studies suggest that IL-12/type I IFN may enhance T cell survival or early proliferation, the mechanisms underlying optimal accumulation of CD8 T cells in vivo are unknown. In particular, it is unclear if disparate signal 3 cytokines optimize effector CD8 T cell accumulation by the same mechanism and how these inflammatory cytokines, which are transiently produced early after infection, affect T cell accumulation many days later at the peak of the immune response. Here, we show that transient exposure of CD8 T cells to IL-12 or type I IFN does not promote survival or confer an early proliferative advantage in vivo, but rather sustains surface expression of CD25, the high-affinity IL-2 receptor. This prolongs division of CD8 T cells in response to basal IL-2, through activation of the PI3K pathway and expression of FoxM1, a positive regulator of cell cycle progression genes. Thus, signal 3 cytokines use a common pathway to optimize effector CD8 T cell accumulation through a temporally orchestrated sequence of cytokine signals that sustain division rather than survival.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Interferon Tipo I/imunologia , Interleucina-12/imunologia , Interleucina-2/metabolismo , Ativação Linfocitária/imunologia , Transdução de Sinais/imunologia , Transferência Adotiva , Animais , Bromodesoxiuridina , Proliferação de Células , Sobrevivência Celular/imunologia , Primers do DNA/genética , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/metabolismo , Immunoblotting , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Análise em Microsséries , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
J Immunol ; 192(1): 35-9, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24285839

RESUMO

During reinfection, high-affinity IgG Abs form complexes with both soluble Ag and Ag displayed on the surface of infected cells. These interactions regulate cellular activation of both innate cells and B cells, which express specific combinations of activating FcγRs (FcγRI, FcγRIII, FcγRIV) and/or the inhibitory FcγR (FcγRIIB). Direct proof for functional expression of FcγR by Ag-specific CD8 T cells is lacking. In this article, we show that the majority of memory CD8 T cells generated by bacterial or viral infection express only FcγRIIB, and that FcγRIIB could be detected on previously activated human CD8 T cells. Of note, FcγR stimulation during in vivo Ag challenge not only inhibited the cytotoxicity of memory CD8 T cells against peptide-loaded or virus-infected targets, but FcγRIIB blockade during homologous virus challenge enhanced the secondary CD8 T cell response. Thus, memory CD8 T cells intrinsically express a functional FcγRIIB, permitting Ag-Ab complexes to regulate secondary CD8 T cell responses.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Regulação da Expressão Gênica , Memória Imunológica/genética , Receptores de IgG/genética , Animais , Antígenos/imunologia , Linhagem Celular , Citotoxicidade Imunológica , Epitopos de Linfócito T/imunologia , Humanos , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Knockout , Receptores de IgG/metabolismo
8.
Immunotherapy ; 3(10): 1223-33, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21995573

RESUMO

Trafficking of CD8 T cells, in both the steady-state and during episodes of infection or inflammation, is a highly dynamic process and involves a variety of receptor-ligand interactions. A thorough, mechanistic understanding of how this process is regulated could potentially lead to disease prevention strategies, through either enhancing (for infectious diseases or tumors) or limiting (for autoimmunity) recruitment of antigen-specific CD8 T cells to areas of tissue inflammation. As CD8 T cells transition from naive to effector to memory cells, changes in gene expression will ultimately dictate anatomical localization of these cells in vivo. In this article, we discuss recent advances in understanding how antigenic stimulation influences expression of various trafficking receptors and ligands, and how this determines the tissue localization of CD8 T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Movimento Celular , Receptores de Retorno de Linfócitos/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Antígenos/imunologia , Movimento Celular/imunologia , Citotoxicidade Imunológica , Regulação da Expressão Gênica/imunologia , Humanos , Memória Imunológica , Inflamação , Ativação Linfocitária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA