Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
J Virol ; 97(1): e0143022, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36598199

RESUMEN

Bovine leukemia virus (BLV) is a retrovirus that causes enzootic bovine leukosis (EBL) in cattle and is widespread in many countries, including Japan. Recent studies have revealed that the expression of immunoinhibitory molecules, such as programmed death-1 (PD-1) and PD-ligand 1, plays a critical role in immunosuppression and disease progression during BLV infection. In addition, a preliminary study has suggested that another immunoinhibitory molecule, T-cell immunoglobulin domain and mucin domain-3 (TIM-3), is involved in immunosuppression during BLV infection. Therefore, this study was designed to further elucidate the immunoinhibitory role of immune checkpoint molecules in BLV infection. TIM-3 expression was upregulated on peripheral CD4+ and CD8+ T cells in BLV-infected cattle. Interestingly, in EBL cattle, CD4+ and CD8+ T cells infiltrating lymphomas expressed TIM-3. TIM-3 and PD-1 were upregulated and coexpressed in peripheral CD4+ and CD8+ T cells from BLV-infected cattle. Blockade by anti-bovine TIM-3 monoclonal antibody increased CD69 expression on T cells and gamma interferon (IFN-γ) production from peripheral blood mononuclear cells from BLV-infected cattle. A syncytium formation assay also demonstrated the antiviral effects of TIM-3 blockade against BLV infection. The combined inhibition of TIM-3 and PD-1 pathways significantly enhanced IFN-γ production and antiviral efficacy compared to inhibition alone. In conclusion, the combined blockade of TIM-3 and PD-1 pathways shows strong immune activation and antiviral effects and has potential as a novel therapeutic method for BLV infection. IMPORTANCE Enzootic bovine leukosis caused by bovine leukemia virus (BLV) is an important viral disease in cattle, causing severe economic losses to the cattle industry worldwide. The molecular mechanisms of BLV-host interactions are complex. Previously, it was found that immune checkpoint molecules, such as PD-1, suppress BLV-specific Th1 responses as the disease progresses. To date, most studies have focused only on how PD-1 facilitates escape from host immunity in BLV-infected cattle and the antiviral effects of the PD-1 blockade. In contrast, how T-cell immunoglobulin domain and mucin domain-3 (TIM-3), another immune checkpoint molecule, regulates anti-BLV immune responses is rarely reported. It is also unclear why PD-1 inhibition alone was insufficient to exert anti-BLV effects in previous clinical studies. In this study, the expression profile of TIM-3 in T cells derived from BLV-infected cattle suggested that TIM-3 upregulation is a cause of immunosuppression in infected cattle. Based on these results, anti-TIM-3 antibody was used to experimentally evaluate its function in influencing immunity against BLV. Results indicated that TIM-3 upregulation induced by BLV infection suppressed T-cell activation and antiviral cytokine production. Some T cells coexpressed PD-1 and TIM-3, indicating that simultaneous inhibition of PD-1 and TIM-3 with their respective antibodies synergistically restored antiviral immunity. This study could open new avenues for treating bovine chronic infections.


Asunto(s)
Leucosis Bovina Enzoótica , Proteínas de Punto de Control Inmunitario , Virus de la Leucemia Bovina , Animales , Bovinos , Linfocitos T CD8-positivos/inmunología , Leucosis Bovina Enzoótica/inmunología , Proteínas de Punto de Control Inmunitario/inmunología , Interferón gamma/inmunología , Virus de la Leucemia Bovina/inmunología , Mucinas/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Regulación de la Expresión Génica/inmunología
2.
Trends Immunol ; 41(8): 692-705, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32605801

RESUMEN

Inhibitory and stimulatory immune checkpoint molecules play important roles in regulating immune responses. An increasing number of these immune regulators are currently being evaluated as targets in putative anti-cancer therapies. Recently, sophisticated genetic screens have been performed to increase our understanding of immune checkpoint pathways and their immunomodulatory regulators. Here, we summarize novel insights obtained by these screens and discuss new directions to advance possible strategies to treat malignancies.


Asunto(s)
Pruebas Genéticas , Proteínas de Punto de Control Inmunitario , Humanos , Proteínas de Punto de Control Inmunitario/genética , Proteínas de Punto de Control Inmunitario/inmunología , Inmunoterapia/tendencias
3.
Br J Cancer ; 126(1): 10-23, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34400801

RESUMEN

Liver cancer is one of the most prevalent cancers, and the third most common cause of cancer-related mortality worldwide. The therapeutic options for the main types of primary liver cancer-hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA)-are very limited. HCC and CCA are immunogenic cancers, but effective immune-mediated tumour control is prevented by their immunosuppressive tumour microenvironment. Despite the critical involvement of key co-inhibitory immune checkpoint interactions in immunosuppression in liver cancer, only a minority of patients with HCC respond to monotherapy using approved checkpoint inhibitor antibodies. To develop effective (combinatorial) therapeutic immune checkpoint strategies for liver cancer, in-depth knowledge of the different mechanisms that contribute to intratumoral immunosuppression is needed. Here, we review the co-inhibitory pathways that are known to suppress intratumoral T cells in HCC and CCA. We provide a detailed description of insights from preclinical studies in cellular crosstalk within the tumour microenvironment that results in interactions between co-inhibitory receptors on different T-cell subsets and their ligands on other cell types, including tumour cells. We suggest alternative immune checkpoints as promising targets, and draw attention to the possibility of combined targeting of co-inhibitory and co-stimulatory pathways to abrogate immunosuppression.


Asunto(s)
Colangiocarcinoma/inmunología , Proteínas de Punto de Control Inmunitario/inmunología , Terapia de Inmunosupresión/métodos , Inmunoterapia/métodos , Neoplasias Hepáticas/patología , Microambiente Tumoral , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Neoplasias de los Conductos Biliares/inmunología , Neoplasias de los Conductos Biliares/patología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/patología , Ensayos Clínicos como Asunto , Humanos , Proteínas de Punto de Control Inmunitario/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/inmunología
4.
Cell Mol Life Sci ; 78(2): 661-673, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32328671

RESUMEN

This study shows that melanoma-associated fibroblasts (MAFs) suppress cytotoxic T lymphocyte (CTL) activity and reveals a pivotal role played by arginase in this phenomenon. MAFs and normal dermal fibroblasts (DFs) were isolated from surgically resected melanomas and identified as Melan-A-/gp100-/FAP+ cells. CTLs of healthy blood donors were activated in the presence of MAF- and DF-conditioned media (CM). Markers of successful CTL activation, cytotoxic degranulation, killing activity and immune checkpoint regulation were evaluated by flow cytometry, ELISPOT, and redirected killing assays. Soluble mediators responsible for MAF-mediated effects were identified by ELISA, flow cytometry, inhibitor assays, and knock-in experiments. In the presence of MAF-CM, activated/non-naïve CTLs displayed dysregulated ERK1/2 and NF-κB signaling, impeded CD69 and granzyme B production, impaired killing activity, and upregulated expression of the negative immune checkpoint receptors TIGIT and BTLA. Compared to DFs, MAFs displayed increased amounts of VISTA and HVEM, a known ligand of BTLA on T cells, increased L-arginase activity and CXCL12 release. Transgenic arginase over-expression further increased, while selective arginase inhibition neutralized MAF-induced TIGIT and BTLA expression on CTLs. Our data indicate that MAF interfere with intracellular CTL signaling via soluble mediators leading to CTL anergy and modify immune checkpoint receptor availability via L-arginine depletion.


Asunto(s)
Arginasa/inmunología , Linfocitos T CD8-positivos/inmunología , Fibroblastos Asociados al Cáncer/inmunología , Proteínas de Punto de Control Inmunitario/inmunología , Melanoma/inmunología , Neoplasias Cutáneas/inmunología , Arginasa/genética , Linfocitos T CD8-positivos/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Células Cultivadas , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de Punto de Control Inmunitario/genética , Activación de Linfocitos , Melanoma/genética , Neoplasias Cutáneas/genética , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
5.
Hum Genet ; 140(6): 865-877, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33452914

RESUMEN

To further explore genetic links between complex traits, we developed a comprehensive framework to harmonize and integrate extensive genotype and phenotype data from the four well-characterized cohorts with the focus on cardiometabolic diseases deposited to the database of Genotypes and Phenotypes (dbGaP). We generated a series of polygenic risk scores (PRS) to investigate pleiotropic effects of loci that confer genetic risk for 19 common diseases and traits on body height, type 2 diabetes (T2D), and myocardial infarction (MI). In a meta-analysis of 20,021 subjects, we identified shared genetic determinants of Crohn's Disease (CD), a type of inflammatory bowel disease, and body height (p = 5.5 × 10-5). The association of PRS-CD with height was replicated in UK Biobank (p = 1.1 × 10-5) and an independent cohort of 510 CD cases and controls (1.57 cm shorter height per PRS-CD interquartile increase, p = 5.0 × 10-3 and a 28% reduction in CD risk per interquartile increase in PRS-height, p = 1.1 × 10-3, with the effect independent of CD diagnosis). A pathway analysis of the variants overlapping between PRS-height and PRS-CD detected significant enrichment of genes from the inflammatory, immune-mediated and growth factor regulation pathways. This finding supports the clinical observation of growth failure in patients with childhood-onset CD and demonstrates the value of using individual-level data from dbGaP in searching for shared genetic determinants. This information can help provide a refined insight into disease pathogenesis and may have major implications for novel therapies and drug repurposing.


Asunto(s)
Estatura/genética , Enfermedad de Crohn/genética , Diabetes Mellitus Tipo 2/genética , Predisposición Genética a la Enfermedad , Péptidos y Proteínas de Señalización Intercelular/genética , Infarto del Miocardio/genética , Adulto , Estatura/inmunología , Niño , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/patología , Bases de Datos Genéticas , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/patología , Femenino , Regulación de la Expresión Génica , Humanos , Proteínas de Punto de Control Inmunitario/genética , Proteínas de Punto de Control Inmunitario/inmunología , Inmunidad Innata , Péptidos y Proteínas de Señalización Intercelular/inmunología , Masculino , Herencia Multifactorial/inmunología , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Fenotipo , Factores de Riesgo
6.
IUBMB Life ; 73(5): 726-738, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33686787

RESUMEN

The importance of the tumor microenvironment in cancer progression has been well studied for many years. Immune checkpoint inhibitors (ICIs) are regarded as potential strategies in enhancing the immune responses in patients with cancer, particularly colorectal cancer (CRC). Notably, CRCs are extraordinarily heterogeneous and mostly are microsatellite-stable (MSS) or cold tumors, which means that the immune response is not usually as strong as that of foreign cells. T-cell immunoglobulin and ITIM domain (TIGIT) is a new immune checkpoint receptor overexpressed inside the CRC tumor-immune microenvironments. Moreover, several studies have shown that TIGIT in combination with other ICIs and/or conventional treatments, can lead to a robust anti-tumor response in CRC. This review looks deep inside TIGIT expression patterns, their various functions, and possible immunotherapy strategies to increase survival rates and decrease immune-related adverse events.


Asunto(s)
Adenocarcinoma/terapia , Neoplasias Colorrectales/terapia , Inhibidores de Puntos de Control Inmunológico , Proteínas de Punto de Control Inmunitario/inmunología , Inmunoterapia/métodos , Receptores Inmunológicos/antagonistas & inhibidores , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Secuencias de Aminoácidos , Animales , Antígenos CD/inmunología , Sistemas CRISPR-Cas , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/radioterapia , Terapia Combinada , Microbioma Gastrointestinal , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Pronóstico , Dominios Proteicos , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Microambiente Tumoral
7.
Gynecol Oncol ; 160(1): 279-284, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33162175

RESUMEN

BACKGROUND: The disappointing activity of single agent immune-checkpoint inhibitors in epitherlial ovarian cancer (EOC) has been attributed in part to its unique tumor microenvironment (TME). IDO, PDL1, LAG3 and TIM3 have been implicated in the immunotolerance of EOC. We investigated the expression of these co-regulators, their change with neoadjuvant chemotherapy (NACT), and their association with outcome. METHOD: We identified 98 patients with EOC treated with NACT and performed IDO, PDL1, LAG3 and TIM3 immunohistochemistry on samples obtained before and after NACT. The cut-off threshold to consider a positive sample was set at 5%. RESULTS: In our cohort, TIM3 was the most prevalent co-regulator, with more than 75% of the samples being TIM3 positive. In comparison, only 22%, 28% and 17% of the samples were considered IDO, PDL1 and LAG3 positive. More than half of ovarian tumors expressed 2, 3 or even all 4 co-inhibitory molecules. However, biomarkers were not correlated with each other. NACT had a marked impact on immune co-regulator expression with over 70% of patients showing a change in biomarker status from negative to positive or vice versa. There was no significant difference in the pattern of co-regulator expression between platinum-sensitive and resistant patients. Co-expression of multiple inhibitory molecules did not appear to affect overall and progression-free survival. CONCLUSION: TIM3 is the most abundant co-inhibitory molecule in OC and may represent an attractive target. In addition, OC frequently co-expressed 2 or more markers supporting ICI combinatorial approaches. Finally, NACT significantly altered the expression of immunosuppressive molecules suggesting that the choice of ICI combinations should be adapted to the composition of the post-NACT immune TME.


Asunto(s)
Proteínas de Punto de Control Inmunitario/biosíntesis , Neoplasias Ováricas/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/biosíntesis , Antígenos CD/inmunología , Antígeno B7-H1/biosíntesis , Antígeno B7-H1/inmunología , Carcinoma Epitelial de Ovario/inmunología , Femenino , Receptor 2 Celular del Virus de la Hepatitis A/biosíntesis , Receptor 2 Celular del Virus de la Hepatitis A/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Proteínas de Punto de Control Inmunitario/inmunología , Inmunohistoquímica , Indolamina-Pirrol 2,3,-Dioxigenasa/biosíntesis , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Persona de Mediana Edad , Terapia Neoadyuvante , Estudios Retrospectivos , Microambiente Tumoral/inmunología , Adulto Joven , Proteína del Gen 3 de Activación de Linfocitos
8.
Curr Rheumatol Rep ; 23(4): 22, 2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33651184

RESUMEN

PURPOSE OF REVIEW: This review will focus on the most common co-inhibitory molecules, emphasizing the importance of these in relation to rheumatic disease. RECENT FINDINGS: Checkpoint molecules are pivotal in determining the outcome of antigen activation. Checkpoint molecules consist of co-stimulatory and co-inhibitory molecules, where the first activates and the latter inhibits the antigen presentation process. Studies show that increased activity of co-inhibitory molecules is associated with a good prognosis in rheumatic diseases. Opposite, when cancer patients are treated with antibodies blocking the inhibitory pathways, autoimmune diseases, including arthritis, develop as immune-related adverse events (IrAE). This emphasizes the importance of these pathways in autoimmune disease. Co-inhibitory molecules are becoming increasingly interesting as future treatment targets in rheumatic conditions. Treatments with antibodies blocking these pathways result in IrAE, often manifesting as autoimmune rheumatic diseases. Therefore, a need to get acquainted with these molecules is growing so we can cope with future challenges in rheumatic diseases.


Asunto(s)
Enfermedades Autoinmunes , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Proteínas de Punto de Control Inmunitario/inmunología , Enfermedades Reumáticas , Artritis , Humanos , Neoplasias , Enfermedades Reumáticas/tratamiento farmacológico , Reumatología
9.
Curr Oncol Rep ; 23(10): 116, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34342752

RESUMEN

PURPOSE OF REVIEW: For patients with metastatic melanoma, immune checkpoint inhibition has drastically changed outcomes. Here, we review the current and next generations of immune-based anti-cancer therapeutics for patients with metastatic melanoma. RECENT FINDINGS: The need for new anti-cancer therapeutics in patients with metastatic melanoma who have progression of disease despite immune checkpoint blockade is evident. Several novel agents are expected to have FDA approval within the next few years, as they have yielded impressive responses. Despite these optimistic agents, the field of immuno-oncology continues to expand and produce agents with novel mechanisms of action. The next generation of immunotherapy is based upon years of thoroughly researched immuno-oncology. Many of these agents are currently being evaluated in early phase clinical trials, and much of the preliminary data looks promising.


Asunto(s)
Inmunoterapia/tendencias , Melanoma/terapia , Tratamiento Basado en Trasplante de Células y Tejidos , Citocinas/inmunología , Citocinas/uso terapéutico , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Proteínas de Punto de Control Inmunitario/inmunología , Inmunidad Innata , Melanoma/inmunología , Melanoma/patología , Linfocitos T/inmunología , Linfocitos T/trasplante , Receptores Toll-Like/agonistas , Receptores Toll-Like/inmunología
10.
Int J Mol Sci ; 22(5)2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33800752

RESUMEN

The members of the B7 family, as immune checkpoint molecules, can substantially regulate immune responses. Since microRNAs (miRs) can regulate gene expression post-transcriptionally, we conducted a scoping review to summarize and discuss the regulatory cross-talk between miRs and new B7 family immune checkpoint molecules, i.e., B7-H3, B7-H4, B7-H5, butyrophilin like 2 (BTNL2), B7-H6, B7-H7, and immunoglobulin like domain containing receptor 2 (ILDR2). The current study was performed using a six-stage methodology structure and Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline. PubMed, Embase, Scopus, Cochrane, ProQuest, and Google Scholar were systematically searched to obtain the relevant records to 5 November 2020. Two authors independently reviewed the obtained records and extracted the desired data. After quantitative and qualitative analyses, we used bioinformatics approaches to extend our knowledge about the regulatory cross-talk between miRs and the abovementioned B7 family members. Twenty-seven articles were identified that fulfilled the inclusion criteria. Studies with different designs reported gene-miR regulatory axes in various cancer and non-cancer diseases. The regulatory cross-talk between the aforementioned B7 family molecules and miRs might provide valuable insights into the pathogenesis of various human diseases.


Asunto(s)
Antígenos B7/fisiología , Proteínas de Punto de Control Inmunitario/fisiología , MicroARNs/fisiología , Antígenos B7/genética , Antígenos B7/inmunología , Coagulación Sanguínea , Quimiocinas/fisiología , Activación de Complemento , Biología Computacional/métodos , Enfermedad/genética , Redes Reguladoras de Genes , Humanos , Proteínas de Punto de Control Inmunitario/genética , Proteínas de Punto de Control Inmunitario/inmunología , MicroARNs/genética , Transducción de Señal
11.
J Cell Physiol ; 235(12): 9834-9850, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32452055

RESUMEN

Renal clear cell carcinoma (ccRCC) is the most common type of renal cell carcinoma, which has strong immunogenicity. A comprehensive study of the role of immune-related genes (IRGs) in ccRCC is of great significance in finding ccRCC treatment targets and improving patient prognosis. In this study, we comprehensively analyzed the expression of IRGs in ccRCC based on The Cancer Genome Atlas datasets. The mechanism of differentially expressed IRGs in ccRCC was analyzed by bioinformatics. In addition, Cox regression analysis was used to screen prognostic related IRGs from differentially expressed IRGs. We also identified a four IRGs signature consisting of four IRGs (CXCL2, SEMA3G, PDGFD, and UCN) through lasso regression and multivariate Cox regression analysis. Further analysis results showed that the four IRGs signature could effectively predict the prognosis of patients with ccRCC, and its predictive power is independent of other clinical factors. In addition, the correlation analysis of immune cell infiltration showed that this four IRGs signature could effectively reflect the level of immune cell infiltration of ccRCC. We also found that the expression of immune checkpoint genes CTLA-4, LAG3, and PD-1 in the high-risk group was higher than that in the low-risk group. Our research revealed the role of IRGs in ccRCC, and developed a four IRGs signature that could be used to evaluate the prognosis of patients with ccRCC, which will help to develop personalized treatment strategies for patients with ccRCC and improve their prognosis. In addition, these four IRGs may be effective therapeutic targets for ccRCC.


Asunto(s)
Carcinoma de Células Renales/inmunología , Quimiocina CXCL2/genética , Linfocinas/genética , Factor de Crecimiento Derivado de Plaquetas/genética , Semaforinas/genética , Urocortinas/genética , Adulto , Anciano , Biomarcadores de Tumor/inmunología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Quimiocina CXCL2/inmunología , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Genoma Humano/inmunología , Humanos , Proteínas de Punto de Control Inmunitario/genética , Proteínas de Punto de Control Inmunitario/inmunología , Inmunogenética , Linfocinas/inmunología , Masculino , Persona de Mediana Edad , Factor de Crecimiento Derivado de Plaquetas/inmunología , Pronóstico , Modelos de Riesgos Proporcionales , Semaforinas/inmunología , Transcriptoma , Urocortinas/inmunología
12.
Br J Cancer ; 122(6): 823-834, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31959920

RESUMEN

BACKGROUND: Immune checkpoint blockade (ICB) therapy improved the prognosis of cancer patients, but general administration of ICBs occasionally induces side effects that include immune-related adverse events and tumour hyper-progression. Here, we established a protein-based system, by which endogenous expression of IC molecule in natural killer (NK) cells was transiently repressed on enhancement of their antitumour activity. METHODS: A protein-based genome modulator (GM) system is composed of a transcription activator-like effector (TALE), DNA methyltransferase and a newly identified potent cell-penetrating peptide with nuclear-trafficking property named NTP. TALE was designed to target the promoter region of the programmed cell death-1 (PD-1) gene. After culturing human NK cells in the presence of NTP-GM protein, we examined endogenous PD-1 expression and antitumour activity of the treated cells. RESULTS: NTP-GM protein efficiently downregulated PD-1 expression in NK cells with increased CpG DNA methylation in the promoter region. The antitumour activity of the treated NK cells was enhanced, and repeated intraperitoneal administrations of the treated NK cells attenuated tumour growth of programmed death-ligand 1-positive tumour cells in vivo. CONCLUSIONS: Because the incorporated NTP-GM protein was quickly degraded and negligible in the administered NK cells, the NTP-GM system could be an alternative option of an ICB without side effects.


Asunto(s)
Represión Epigenética/inmunología , Proteínas de Punto de Control Inmunitario/inmunología , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Animales , Humanos , Ratones , Pronóstico
13.
Protein Expr Purif ; 170: 105596, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32036001

RESUMEN

Antibodies that block interaction of immune checkpoint receptors with its ligands have revolutionized the treatment of several cancers. Despite the success of this approach, the high cost has been restricted the use of this class of drugs. In this context, the development of biosimilar can be an important strategy for reducing prices and expanding access after patent has been dropped. Here, we evaluated the use of HEK293 cells for transient expression of an immune checkpoint-blocking antibody as a first step for biosimilar development. Antibody light and heavy chain genes were cloned into pCI-neo vector and transiently expressed in HEK293 cells. The culture supernatant was then subjected to protein A affinity chromatography, which allowed to obtain the antibody with high homogeneity. For physicochemical comparability, biosimilar antibody and reference drug were analyzed by SDS-PAGE, isoelectric focusing, circular dichroism and fluorescence spectroscopy. The results indicated that the both antibodies have a high degree of structural similarity. Lastly, the biosimilar antibody binding capacity to target receptor was shown to be similar to reference product in ELISA and flow cytometry assays. These data demonstrate that the HEK293 system can be used as an important tool for candidate selection and early development of biosimilar antibodies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Biosimilares Farmacéuticos/farmacología , Inhibidores de Puntos de Control Inmunológico/farmacología , Proteínas de Punto de Control Inmunitario/genética , Cadenas Pesadas de Inmunoglobulina/farmacología , Cadenas Ligeras de Inmunoglobulina/farmacología , Anticuerpos Monoclonales/biosíntesis , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Biosimilares Farmacéuticos/metabolismo , Cromatografía de Afinidad , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Células HEK293 , Humanos , Inhibidores de Puntos de Control Inmunológico/inmunología , Proteínas de Punto de Control Inmunitario/inmunología , Cadenas Pesadas de Inmunoglobulina/biosíntesis , Cadenas Ligeras de Inmunoglobulina/biosíntesis , Focalización Isoeléctrica
14.
Int J Mol Sci ; 21(21)2020 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-33167514

RESUMEN

Although the ever-increasing number of cancer patients pose substantial challenges worldwide, finding a treatment with the highest response rate and the lowest number of side effects is still undergoing research. Compared to chemotherapy, the relatively low side effects of cancer immunotherapy have provided ample opportunity for immunotherapy to become a promising approach for patients with malignancy. However, the clinical translation of immune-based therapies requires robust anti-tumoral immune responses. Immune checkpoints have substantial roles in the induction of an immunosuppressive tumor microenvironment and tolerance against tumor antigens. Identifying and targeting these inhibitory axes, which can be established between tumor cells and tumor-infiltrating lymphocytes, can facilitate the development of anti-tumoral immune responses. Bispecific T-cell engagers, which can attract lymphocytes to the tumor microenvironment, have also paved the road for immunological-based tumor elimination. The development of CAR-T cells and their gene editing have brought ample opportunity to recognize tumor antigens, independent from immune checkpoints and the major histocompatibility complex (MHC). Indeed, there have been remarkable advances in developing various CAR-T cells to target tumoral cells. Knockout of immune checkpoints via gene editing in CAR-T cells might be designated for a breakthrough for patients with malignancy. In the midst of this fast progress in cancer immunotherapies, there is a need to provide up-to-date information regarding immune checkpoints, bispecific T-cell engagers, and CAR-T cells. Therefore, this review aims to provide recent findings of immune checkpoints, bispecific T-cell engagers, and CAR-T cells in cancer immunotherapy and discuss the pertained clinical trials.


Asunto(s)
Proteínas de Punto de Control Inmunitario/inmunología , Inmunoterapia Adoptiva/métodos , Inmunoterapia Adoptiva/tendencias , Antígenos de Neoplasias/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Proteínas de Punto de Control Inmunitario/metabolismo , Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/fisiopatología , Linfocitos T/inmunología , Microambiente Tumoral/inmunología
15.
Sci Immunol ; 7(67): eabj5501, 2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-35061505

RESUMEN

The interaction of SIRPα with CD47 represents a major mechanism for preventing macrophage phagocytosis. However, CD47-independent mechanisms are poorly defined. Here, we report a critical role of SLAM family receptors (SFRs), ubiquitously expressed on hematopoietic cells and forming homotypic interactions, in constraining macrophage phagocytosis. We found that SFR deficiency triggered macrophage phagocytosis of hematopoietic cells, leading to severe rejection of donor hematopoietic graft in recipient mice. Specific SFR members, mainly SLAMF3 and SLAMF4, were identified as "don't eat me" receptors on macrophages. These receptors inhibited "eat me" signals, such as LRP1-mediated activation of mTOR and Syk, through SH2 domain-containing phosphatases. SFRs combined with, but were independent of, CD47 to mitigate macrophage phagocytosis, and the combined deletion of SFRs and CD47 resulted in hematopoietic cytopenia in mice. This SFR-mediated tolerance was compromised in patients with hemophagocytic lymphohistiocytosis, a syndrome characterized by inappropriate phagocytosis toward hematopoietic cells. Loss of SFRs potently elicited macrophage rejection of hematopoietic tumors. Deletion of SFRs also significantly enhanced the phagocytosis of CD19-positive hematopoietic targets by the macrophages expressing the chimeric CD19 antigen receptor. Therefore, SFR-mediated inhibition of macrophage phagocytosis is critical to hematopoietic homeostasis, and SFRs may represent previously unknown targets for tumor immunotherapy.


Asunto(s)
Neoplasias Hematológicas/inmunología , Proteínas de Punto de Control Inmunitario/inmunología , Macrófagos/inmunología , Fagocitosis/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología , Animales , Línea Celular , Neoplasias Hematológicas/patología , Humanos , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
16.
Int Immunopharmacol ; 104: 108512, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35026655

RESUMEN

NLRP3 plays a pathogenic role in tumorigenesis by regulating innate and acquired immunity, apoptosis, differentiation, and intestinal microbes in tumors. Our research aimed to investigate the role of NLRP3 in pan-cancers based on multi-omics data in the TCGA database. Most types of tumors showed increased expression of NLRP3. Among them, the overexpressed NLRP3 in liver hepatocellular carcinoma (LIHC) and ovarian cancer (OV) indicated worse overall survival (OS). Further analysis also confirmed overexpressed NLRP3 in colon cancer (COAD) indicated a high probability of microsatellite instability (MSI) and low tumor mutational burden (TMB), which indicated a better response to immune checkpoint inhibitors (ICIs). Interestingly, overexpression of NLRP3 was closely related to high infiltration of immune cells (T cells, B cells, etc.) and overexpressed immune checkpoints (PD-1, PD-L1, LAG3, etc.). These results demonstrated NLRP3 promoted immune escape in cancers. Finally, we investigated the expression of various immune checkpoints by treating NLRP3 inhibitor MCC950 during the co-culture of peripheral blood mononuclear cells (PBMC) and LIHC cell line Hep3B. MCC950 significantly repressed the expression of PD-L1 and LAG3, and promoted the apoptosis rate of Hep3B. In conclusion, our research demonstrated the role of NLRP3 in pan-cancer, especially in LIHC. Inhibition of NLRP3 promoted the killing effect of T cells to cancer cells by repressing the expression of immune checkpoints.


Asunto(s)
Inflamasomas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Neoplasias/inmunología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Técnicas de Cocultivo , Metilación de ADN , Humanos , Proteínas de Punto de Control Inmunitario/inmunología , Inflamasomas/genética , Estimación de Kaplan-Meier , Leucocitos Mononucleares , Mutación , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Neoplasias/genética , Neoplasias/mortalidad , Escape del Tumor
17.
J Pathol Clin Res ; 8(4): 340-354, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35289116

RESUMEN

The tumor microenvironment (TME) is a critical regulator of the development of malignant lymphoma. Therapeutics targeting the TME, especially immune checkpoint molecules, are changing the treatment strategy for lymphoma. However, the overall response to these therapeutics for diffuse large B-cell lymphoma (DLBCL) is modest and new targets of immunotherapy are needed. To find critical immune checkpoint molecules for DLBCL, we explored the prognostic impact of immune checkpoint molecules and their ligands using publicly available datasets of gene expression profiles. In silico analysis of three independent datasets (GSE117556, GSE10846, and GSE181063) revealed that DLBCL expressing CD24 had a poor prognosis and had a high frequency of MYC aberrations. Moreover, gene set enrichment analysis showed that the 'MYC-targets-hallmark' (false discovery rate [FDR] = 0.024) and 'inflammatory-response-hallmark' (FDR = 0.001) were enriched in CD24-high and CD24-low DLBCL, respectively. In addition, the expression of cell-specific markers of various immune cells was higher in CD24-low DLBCL than in CD24-high DLBCL. CIBERSORT analysis of the datasets showed fewer macrophages in CD24-high DLBCL than in CD24-low DLBCL. Additionally, immunohistochemical analysis of 335 cases of DLBCL showed that few TME cells were found in CD24-high DLBCL, although statistical differences were not observed. These data indicate that CD24 expression suppresses immune cell components of the TME in DLBCL, suggesting that CD24 may be a target for cancer immunotherapy in aggressive large B-cell lymphoma.


Asunto(s)
Antígeno CD24 , Proteínas de Punto de Control Inmunitario , Linfoma de Células B Grandes Difuso , Antígeno CD24/genética , Antígeno CD24/inmunología , Humanos , Proteínas de Punto de Control Inmunitario/inmunología , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/patología , Fenotipo , Pronóstico , Microambiente Tumoral
18.
Signal Transduct Target Ther ; 6(1): 28, 2021 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-33479196

RESUMEN

The immune system initiates robust immune responses to defend against invading pathogens or tumor cells and protect the body from damage, thus acting as a fortress of the body. However, excessive responses cause detrimental effects, such as inflammation and autoimmune diseases. To balance the immune responses and maintain immune homeostasis, there are immune checkpoints to terminate overwhelmed immune responses. Pathogens and tumor cells can also exploit immune checkpoint pathways to suppress immune responses, thus escaping immune surveillance. As a consequence, therapeutic antibodies that target immune checkpoints have made great breakthroughs, in particular for cancer treatment. While the overall efficacy of immune checkpoint blockade (ICB) is unsatisfactory since only a small group of patients benefited from ICB treatment. Hence, there is a strong need to search for other targets that improve the efficacy of ICB. Ubiquitination is a highly conserved process which participates in numerous biological activities, including innate and adaptive immunity. A growing body of evidence emphasizes the importance of ubiquitination and its reverse process, deubiquitination, on the regulation of immune responses, providing the rational of simultaneous targeting of immune checkpoints and ubiquitination/deubiquitination pathways to enhance the therapeutic efficacy. Our review will summarize the latest findings of ubiquitination/deubiquitination pathways for anti-tumor immunity, and discuss therapeutic significance of targeting ubiquitination/deubiquitination pathways in the future of immunotherapy.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Inmunidad/inmunología , Neoplasias/inmunología , Ubiquitinación/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Proteínas de Punto de Control Inmunitario/inmunología , Inmunoterapia , Inflamación/inmunología
19.
Cells ; 10(5)2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-34068035

RESUMEN

Autoreactive B cells are key drivers of pathogenic processes in autoimmune diseases by the production of autoantibodies, secretion of cytokines, and presentation of autoantigens to T cells. However, the mechanisms that underlie the development of autoreactive B cells are not well understood. Here, we review recent studies leveraging novel techniques to identify and characterize (auto)antigen-specific B cells. The insights gained from such studies pertaining to the mechanisms involved in the escape of tolerance checkpoints and the activation of autoreactive B cells are discussed. In addition, we briefly highlight potential therapeutic strategies to target and eliminate autoreactive B cells in autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Autoinmunidad , Linfocitos B/inmunología , Proteínas de Punto de Control Inmunitario/inmunología , Tolerancia Inmunológica , Activación de Linfocitos , Animales , Enfermedades Autoinmunes/metabolismo , Linfocitos B/metabolismo , Humanos , Proteínas de Punto de Control Inmunitario/metabolismo , Fenotipo
20.
BMB Rep ; 54(8): 403-412, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34078531

RESUMEN

In the tumor microenvironment, immune checkpoint ligands (ICLs) must be expressed in order to trigger the inhibitory signal via immune checkpoint receptors (ICRs). Although ICL expression frequently occurs in a manner intrinsic to tumor cells, extrinsic factors derived from the tumor microenvironment can fine-tune ICL expression by tumor cells or prompt non-tumor cells, including immune cells. Considering the extensive interaction between T cells and other immune cells within the tumor microenvironment, ICL expression on immune cells can be as significant as that of ICLs on tumor cells in promoting antitumor immune responses. Here, we introduce various regulators known to induce or suppress ICL expression in either tumor cells or immune cells, and concise mechanisms relevant to their induction. Finally, we focus on the clinical significance of understanding the mechanisms of ICLs for an optimized immunotherapy for individual cancer patients. [BMB Reports 2021; 54(8): 403-412].


Asunto(s)
Proteínas de Punto de Control Inmunitario/genética , Proteínas de Punto de Control Inmunitario/inmunología , Microambiente Tumoral/inmunología , Expresión Génica/genética , Expresión Génica/inmunología , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inmunoterapia , Ligandos , Neoplasias/patología , Receptores Inmunológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA