Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Cancer Immunol Immunother ; 73(5): 81, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38554184

RESUMEN

Poliovirus receptor-related immunoglobulin domain-containing protein, or PVRIG, is a newly discovered immune checkpoint that has emerged as a promising target for cancer immunotherapy. It is primarily expressed on activated T and natural killer (NK) cells, and once engaged with its ligand, PVRL2, it induces inhibitory signaling in T cells, thereby promoting the functional exhaustion of tumor-infiltrating lymphocytes (TILs). Here, we characterized IBI352g4a, a novel humanized anti-PVRIG antibody with Fc-competent function, explored the mechanism of its antitumor activity in preclinical models, and systemically evaluated the contribution of FcrR engagement to PVRIG blockade-induced antitumor activity. IBI352g4a binds to the extracellular domain of human PVRIG with high affinity (Kd = 0.53 nM) and specificity, and fully blocks the interaction between PVRIG and its ligand PVRL2. Unlike other immune checkpoints, IBI352g4a significantly induced NK cell activation and degranulation, but had a minimal effect on T-cell activation in in vitro functional assays. IBI352g4a induced strong antitumor effect in several preclinic models, through in vivo mechanism analysis we found that both NK and T cells contribute to the antitumor effect, but NK cells play predominant roles. Specifically, a single dose of IBI352g4a induced significant NK cell activation in TILs, but T-cell activation was observed only after the second dose. Moreover, the Fc effector function is critical for both NK cell activation and treatment efficacy in vitro and in vivo. Our study, for the first time, demonstrates that both NK activation and FcrR engagement are required for antitumor efficacy induced by PVRIG blockade.


Asunto(s)
Células Asesinas Naturales , Neoplasias , Humanos , Ligandos , Inmunoterapia , Linfocitos Infiltrantes de Tumor , Neoplasias/metabolismo
2.
Microbiology (Reading) ; 167(1)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33295859

RESUMEN

The CpxRA two-component regulatory system and the Rcs phosphorelay system are both employed by the Enterobacteriaceae family to preserve bacterial envelope integrity and function when growing under stress. Although both systems regulate several overlapping physiological processes, evidence demonstrating a molecular connection between Cpx and Rcs signalling outputs is scarce. Here, we show that CpxR negatively regulates the transcription of the rcsB gene in the Rcs phosphorelay system in Yersinia pseudotuberculosis. Interestingly, transcription of rcsB is under the control of three promoters, which were all repressed by CpxR. Critically, synthetic activation of Cpx signalling through mislocalization of the NlpE lipoprotein to the inner membrane resulted in an active form of CpxR that repressed activity of rcsB promoters. On the other hand, a site-directed mutation of the phosphorylation site at residue 51 in CpxR generated an inactive non-phosphorylated variant that was unable to regulate output from these rcsB promoters. Importantly, CpxR-mediated inhibition of rcsB transcription in turn restricted activation of the Ysc-Yop type III secretion system (T3SS). Moreover, active CpxR blocks zinc-mediated activation of Rcs signalling and the subsequent activation of lcrF transcription. Our results demonstrate a novel regulatory cascade linking CpxR-RcsB-LcrF to control production of the Ysc-Yop T3SS.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Sistemas de Secreción Tipo III/metabolismo , Yersinia pseudotuberculosis/metabolismo , Proteínas Bacterianas/genética , Fosforilación , Regiones Promotoras Genéticas , Sistemas de Secreción Tipo III/genética , Yersinia pseudotuberculosis/genética
3.
Cancer Lett ; 536: 215663, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35381307

RESUMEN

Multiple myeloma (MM) is a hematological malignancy that results from the malignant proliferation of plasma cells in the bone marrow. B cell maturation antigen (BCMA) is highly selectively expressed in malignant plasma cells and is a novel therapeutic target for MM. Here, we developed a bispecific T cell engager, IBI379, that targets BCMA and CD3, and investigated its antitumor efficacy against MM. IBI379 showed strong binding affinity with both BCMA and CD3, which triggered T cell activation, proliferation, and cytokine release. An in vitro study demonstrated that IBI379 induced the lysis of MM cells expressing differing levels of BCMA on the cell surface. Administration of IBI379 in H929 or Daudi-BCMA cell xenograft mouse models significantly inhibited tumor growth without inducing body weight loss. The mechanism of action study revealed the accumulation of CD4+CD8+ T cells and granzyme B-positive T cells in tumors that were treated with IBI379. Moreover, administration of low dose of IBI379 in cynomolgus monkeys was well-tolerated and induced the depletion of BCMA+ B cells and a mild transient increase of cytokines. Collectively, these results demonstrate that IBI379 is a highly potent therapeutic strategy for depleting BCMA-positive B cells and is a promising approach for the treatment of MM.


Asunto(s)
Anticuerpos Biespecíficos , Mieloma Múltiple , Animales , Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/uso terapéutico , Antígeno de Maduración de Linfocitos B/metabolismo , Complejo CD3/inmunología , Linfocitos T CD8-positivos/metabolismo , Citocinas/uso terapéutico , Humanos , Ratones , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
FEMS Microbiol Lett ; 368(6)2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33705525

RESUMEN

LcrF is the master regulator that positively regulates the Ysc type III secretion system (T3SS) in Yersinia and shares a high similarity with the DNA-binding domain of the T3SS master regulator ExsA in Pseudomonas aeruginosa. Based on these features, bioinformatics analysis has predicted a putative LcrF-binding site in its target promoters. Here, we experimentally characterized its binding motif. An adenine-rich LcrF-binding region in the lcrG promoter sequence, a typical regulatory target of LcrF, was first confirmed. To obtain detailed information, this binding region was cloned into a synthetized promoter and mutations in this region were further constructed. We demonstrated that the 5'-AAAAA-n5-GnCT-3' sequence is required for LcrF regulation and this motif is strictly located 4-bp upstream of a noncanonical promoter, in which the -35 and -10 elements are separated by a 21-bp spacer. Consistently, the putative binding motif was found in promoters of nine T3SS related operons or genes positively regulated by LcrF. Transcriptome analysis further confirmed that LcrF specifically activates T3SS genes in Yersinia. Collectively, our data suggest that LcrF has evolved to be a specific T3SS activator with a stringent sequence requirement for transcriptional regulation.


Asunto(s)
Secuencias de Aminoácidos , Proteínas Bacterianas , Transactivadores , Secuencias de Aminoácidos/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Unión Proteica , Transactivadores/química , Transactivadores/metabolismo , Yersinia pseudotuberculosis/química , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/metabolismo
5.
mBio ; 12(3): e0145721, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34154409

RESUMEN

Human-pathogenic Yersinia species employ a plasmid-encoded type III secretion system (T3SS) to negate immune cell function during infection. A critical element in this process is the coordinated regulation of T3SS gene expression, which involves both transcriptional and posttranscriptional mechanisms. LcrQ is one of the earliest identified negative regulators of Yersinia T3SS, but its regulatory mechanism is still unclear. In a previous study, we showed that LcrQ antagonizes the activation role played by the master transcriptional regulator LcrF. In this study, we confirm that LcrQ directly interacts with LcrH, the chaperone of YopD, to facilitate the negative regulatory role of the YopD-LcrH complex in repressing lcrF expression at the posttranscriptional level. Negative regulation is strictly dependent on the YopD-LcrH complex, more so than on LcrQ. The YopD-LcrH complex helps to retain cytoplasmic levels of LcrQ to facilitate the negative regulatory effect. Interestingly, RNase E and its associated protein RhlB participate in this negative regulatory loop through a direct interaction with LcrH and LcrQ. Hence, we present a negative regulatory loop that physically connects LcrQ to the posttranscriptional regulation of LcrF, and this mechanism incorporates RNase E involved in mRNA decay. IMPORTANCE All three human-pathogenic Yesinia species, Y. pestis, Y. enterocolitica, and Y. pseudotuberculosis, employ a plasmid-encoded T3SS to target immunomodulatory effectors into host immune cells. Several plasmid-encoded regulators influence T3SS control, including the master transcriptional activator LcrF, the posttranscriptional repressor YopD, and the unassigned negative regulatory factor LcrQ. Since LcrQ lacks any obvious DNA or RNA binding domains, its regulatory mechanism might be special. In this study, we screened for proteins that directly engaged with LcrQ. We found that LcrQ cooperates with LcrH of the YopD-LcrH complex to aid in the posttranscriptional repression of lcrF expression. This negative-control loop also involved the mRNA decay factor RNase E and its associated RhlB protein, which were recruited to the regulatory complex by both LcrQ and LcrH. Hence, we identify interacting components of LcrQ that shed new light on a mechanism inhibiting T3SS production and biogenesis.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Chaperonas Moleculares/genética , Sistemas de Secreción Tipo III/genética , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/metabolismo , Endorribonucleasas/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Sistemas de Secreción Tipo III/metabolismo
6.
MAbs ; 11(8): 1443-1451, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31402780

RESUMEN

Blockade of immune checkpoint pathways by programmed cell death protein 1 (PD-1) antibodies has demonstrated broad clinical efficacy against a variety of malignancies. Sintilimab, a highly selective, fully human monoclonal antibody (mAb), blocks the interaction of PD-1 and its ligands and has demonstrated clinical benefit in various clinical studies. Here, we evaluated the affinity of sintilimab to human PD-1 by surface plasmon resonance and mesoscale discovery and evaluated PD-1 receptor occupancy and anti-tumor efficacy of sintilimab in a humanized NOD/Shi-scid-IL2rgamma (null) (NOG) mouse model. We also assessed the receptor occupancy and immunogenicity of sintilimab from clinical studies in humans (9 patients with advanced solid tumor and 381 patients from 4 clinical studies, respectively). Sintilimab bound to human PD-1 with greater affinity than nivolumab (Opdivo®, MDX-1106) and pembrolizumab (Keytruda®, MK-3475). The high affinity of sintilimab is explained by its distinct structural binding mode to PD-1. The pharmacokinetic behavior of sintilimab did not show any significant differences compared to the other two anti-PD-1 mAbs. In the humanized NOG mouse model, sintilimab showed superior PD-1 occupancy on circulating T cells and a stronger anti-tumor effect against NCI-H292 tumors. The strong anti-tumor response correlated with increased interferon-γ-secreting, tumor-specific CD8+ T cells, but not with CD4+ Tregs in tumor tissue. Pharmacodynamics testing indicated a sustained mean occupancy of ≥95% of PD-1 molecules on circulating T cells in patients following sintilimab infusion, regardless of infusion dose. Sintilimab infusion was associated with 0.52% (2/381 patients) of anti-drug antibodies and 0.26% (1/381 patients) neutralizing antibodies. These data validate sintilimab as a novel, safe, and efficacious anti-PD-1 mAb for cancer immunotherapy.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Linfocitos T CD8-positivos/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias Experimentales , Receptor de Muerte Celular Programada 1/inmunología , Transducción de Señal/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Linfocitos T CD8-positivos/patología , Línea Celular Tumoral , Humanos , Inmunoterapia , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Transducción de Señal/inmunología , Linfocitos T Reguladores/patología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA