Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Trends Cancer ; 10(7): 584-587, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38839545

RESUMEN

B7-H3, an immune checkpoint glycoprotein, facilitates immune evasion and the promotion of tumors and is highly expressed on the surface of prostate cancer (PCa) cells, which makes it a feasible and robust candidate for immunotherapies against advanced prostate cancer. Here, we summarize and discuss recent findings on the suitability of targeting B7-H3 in PCa treatment.


Asunto(s)
Antígenos B7 , Inmunoterapia , Neoplasias de la Próstata , Humanos , Masculino , Antígenos B7/antagonistas & inhibidores , Antígenos B7/inmunología , Antígenos B7/metabolismo , Antígenos B7/genética , Neoplasias de la Próstata/terapia , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Inmunoterapia/métodos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Animales , Terapia Molecular Dirigida/métodos
2.
Int Immunopharmacol ; 132: 111926, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38552297

RESUMEN

Mortality due to malignant tumors is one of the major factors affecting the life expectancy of the global population. Therapeutic antibodies are a cutting-edge treatment method for restricting tumor growth. B7-H3 is highly expressed in tumor tissues, but rarely in normal tissues. B7-H3 is closely associated with poor prognosis in patients with tumors. B7-H3 is an important target for antitumor therapy. In this study, the fully human anti-B7H3 single-chain antibodies (scFvs) were isolated and screened from the fully human phage immune library with B7H3 as the target. The antibodies screened from a fully human phage library had low immunogenicity and high affinity, which was more beneficial for clinical application. Leveraging B7-H3 scFvs as a foundation, we constructed two distinct recombinant antibody formats, scFv-Fc and IgG1, characterized by elevated affinity and a prolonged half-life. The results demonstrated that the recombinant antibodies had high specificity and affinity for the B7-H3 antigen and inhibited tumor cell growth by enhancing the ADCC. After treatment with anti-B7H3 recombinant antibody, the number of infiltrating T cells in the tumor increased and the secretion of IFN- γ by infiltrating T cells increased in vivo. Additionally, the use of pleural fluid samples obtained from tumor-afflicted patients revealed the ability of anti-B7-H3 recombinant antibodies to reverse CD8+ T cell exhaustion. In summary, we screened the fully human anti-B7H3 recombinant antibodies with specificity and high affinity that increase immune cell infiltration and IFN-γ secretion, thereby inhibiting tumor cell growth to a certain extent. This finding provides a theoretical basis for the development of therapeutic tumor antibodies and could help promote further development of antibody-based drugs.


Asunto(s)
Antígenos B7 , Anticuerpos de Cadena Única , Antígenos B7/inmunología , Antígenos B7/metabolismo , Antígenos B7/genética , Antígenos B7/antagonistas & inhibidores , Humanos , Animales , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/farmacología , Anticuerpos de Cadena Única/uso terapéutico , Línea Celular Tumoral , Ratones , Femenino , Linfocitos T/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/uso terapéutico , Ratones Endogámicos C57BL , Masculino , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Interferón gamma/metabolismo , Interferón gamma/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos
3.
Int J Mol Sci ; 22(19)2021 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-34639059

RESUMEN

Gastric cancer (GC), with a heterogeneous nature, is the third leading cause of death worldwide. Over the past few decades, stable reductions in the incidence of GC have been observed. However, due to the poor response to common treatments and late diagnosis, this cancer is still considered one of the lethal cancers. Emerging methods such as immunotherapy with immune checkpoint inhibitors (ICIs) have transformed the landscape of treatment for GC patients. There are presently eleven known members of the B7 family as immune checkpoint molecules: B7-1 (CD80), B7-2 (CD86), B7-H1 (PD-L1, CD274), B7-DC (PDCD1LG2, PD-L2, CD273), B7-H2 (B7RP1, ICOS-L, CD275), B7-H3 (CD276), B7-H4 (B7x, B7S1, Vtcn1), B7-H5 (VISTA, Gi24, DD1α, Dies1 SISP1), B7-H6 (NCR3LG1), B7-H7 (HHLA2), and Ig-like domain-containing receptor 2 (ILDR2). Interaction of the B7 family of immune-regulatory ligands with the corresponding receptors resulted in the induction and inhibition of T cell responses by sending co-stimulatory and co-inhibitory signals, respectively. Manipulation of the signals provided by the B7 family has significant potential in the management of GC.


Asunto(s)
Antígenos B7/inmunología , Inmunomodulación , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/metabolismo , Animales , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Antígenos B7/antagonistas & inhibidores , Antígenos B7/química , Antígenos B7/genética , Biomarcadores de Tumor , Proteínas Portadoras , Ensayos Clínicos como Asunto , Regulación de la Expresión Génica , Humanos , Inmunomodulación/efectos de los fármacos , Inmunomodulación/genética , Terapia Molecular Dirigida , Familia de Multigenes , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Isoformas de Proteínas , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología , Resultado del Tratamiento
4.
Reprod Biol Endocrinol ; 19(1): 114, 2021 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-34289871

RESUMEN

BACKGROUND: The immune mechanism was shown to be involved in the development of adenomyosis. The aim of the current study was to evaluate the expression of the immune checkpoints B7-H2, B7-H3, B7-H4 and PD-L2 in adenomyosis and to explore the effect of mifepristone on the expression of these immune checkpoints. METHODS: The expression of B7-H2, B7-H3, B7-H4 and PD-L2 in normal endometria and adenomyosis patient samples treated with or without mifepristone was determined by immunohistochemistry analysis. RESULTS: In adenomyosis patient samples, the expression of B7-H2, B7-H3 and B7-H4 was increased in the eutopic and ectopic endometria compared with normal endometria, both in the proliferative and secretory phases. Moreover, the expression of B7-H2 and B7-H3 was higher in adenomyotic lesions than in the corresponding eutopic endometria, both in the proliferative and secretory phases. The expression of PD-L2 was higher in adenomyotic lesions than in normal endometria in both the proliferative and secretory phases. In the secretory phase but not the proliferative phase, the expression of B7-H4 and PD-L2 in adenomyotic lesions was significantly higher than that in the corresponding eutopic endometria. In normal endometria and eutopic endometria, the expression of B7-H4 was elevated in the proliferative phase compared with that in the secretory phase, while in the ectopic endometria, B7-H4 expression was decreased in the proliferative phase compared with the secretory phase. In addition, the expression of B7-H2, B7-H3, B7-H4 and PD-L2 was significantly decreased in adenomyosis tissues after treatment with mifepristone. CONCLUSIONS: The expression of the immune checkpoint proteins B7-H2, B7-H3, B7-H4 and PD-L2 is upregulated in adenomyosis tissues and is downregulated with mifepristone treatment. The data suggest that B7 immunomodulatory molecules are involved in the pathophysiology of adenomyosis.


Asunto(s)
Adenomiosis/metabolismo , Antígenos B7/biosíntesis , Ligando Coestimulador de Linfocitos T Inducibles/biosíntesis , Mifepristona/uso terapéutico , Proteína 2 Ligando de Muerte Celular Programada 1/biosíntesis , Inhibidor 1 de la Activación de Células T con Dominio V-Set/biosíntesis , Adenomiosis/tratamiento farmacológico , Adenomiosis/genética , Adulto , Antígenos B7/antagonistas & inhibidores , Antígenos B7/genética , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Femenino , Expresión Génica , Antagonistas de Hormonas/farmacología , Antagonistas de Hormonas/uso terapéutico , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Ligando Coestimulador de Linfocitos T Inducibles/antagonistas & inhibidores , Ligando Coestimulador de Linfocitos T Inducibles/genética , Persona de Mediana Edad , Mifepristona/farmacología , Proteína 2 Ligando de Muerte Celular Programada 1/antagonistas & inhibidores , Proteína 2 Ligando de Muerte Celular Programada 1/genética , Inhibidor 1 de la Activación de Células T con Dominio V-Set/antagonistas & inhibidores , Inhibidor 1 de la Activación de Células T con Dominio V-Set/genética
5.
Front Immunol ; 12: 676181, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093577

RESUMEN

Immune checkpoints (ICs) have pivotal roles in regulating immune responses. The inhibitory ICs in the tumor microenvironment (TME) have been implicated in the immune evasion of tumoral cells. Therefore, identifying and targeting these inhibitory ICs might be critical for eliminating tumoral cells. V-domain immunoglobulin suppressor of T cell activation (VISTA) is a novel inhibitory IC that is expressed on myeloid cells, lymphoid cells, and tumoral cells; therefore, VISTA can substantially regulate innate and adaptive anti-tumoral immune responses. Besides, growing evidence indicates that VISTA blockade can enhance the sensitivity of tumoral cells to conventional IC-based immunotherapy, e.g., cytotoxic T lymphocyte antigen 4 (CTLA-4) inhibitors. In this regard, the current study aimed to review the current evidence about the structure and expression pattern of VISTA, its role in TME, the clinicopathological significance of VISTA, and its prognostic values in various cancers. Besides, this review intended to collect the lessons from the recent pre-clinical and clinical studies and propose a strategy to overcome tumor immune-resistance states.


Asunto(s)
Antígenos B7/fisiología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/tratamiento farmacológico , Antígenos B7/antagonistas & inhibidores , Antígenos B7/química , Ensayos Clínicos como Asunto , Humanos , Microambiente Tumoral
6.
Invest New Drugs ; 39(5): 1267-1274, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33909231

RESUMEN

The families of miR-34 and miR-449 share the same seed region. However, the members showed differential effects on the expression of B7-H3 and PD-L1 in HCT-116 cells. Using miR-34a as a template, the non-seed region was modified by nucleotide alteration, yielding four synthetic microRNA (miRNA) analogs. Among those, NS-MX3, with a base alteration from G to C at the 18th locus of miR-34a, showed the most potent inhibition on both B7-H3 and PD-L1 expression. Subsequent investigations demonstrated that NS-MX3 had a broad anti-proliferation activity against several colorectal tumor cell lines and its antitumor effect was consistently reflected by tumor growth inhibition (TGI) in the HCT-116 xenograft model. In addition, NS-MX3 displayed a synergistic effect on TGI when combined with bevacizumab or regorafenib. Further analysis revealed that the superior antitumor activity of NS-MX3 was correlated to concomitant suppression of both B7-H3 and PD-L1 expression in tumor tissues. Taken together, the present study indicates that the non-seed region of miRNAs plays an important role in the regulation of checkpoint genes, thus showcasing single nucleotide alteration of the non-seed region as a promising approach to discover and develop novel immunotherapies.


Asunto(s)
Antineoplásicos/farmacología , Antígenos B7/antagonistas & inhibidores , Neoplasias Colorrectales/patología , MicroARNs/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Bevacizumab/uso terapéutico , Biomarcadores de Tumor , Línea Celular Tumoral , Humanos , Masculino , Ratones SCID , Compuestos de Fenilurea/uso terapéutico , Piridinas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cell Biochem Biophys ; 79(2): 397-405, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33743142

RESUMEN

BACKGROUND: B7 homolog 3 (B7-H3), a member of the immunoregulatory ligand B7 family, is pivotal in T-cell-mediated immune response. It is widely expressed in diverse human tumors and its high expression indicates the poor prognosis of the patients. Nonetheless, B7-H3's role in colorectal cancer (CRC) needs to be further explored. METHODS: Western blot and immunohistochemistry were employed for detecting B7-H3 protein expression in CRC tissues and cell lines, respectively. Quantitative real-time polymerase chain reaction (qRT-PCR) was utilized for detecting B7-H3 mRNA and miR-128 expression levels. CRC cell lines SW620 and HT29 were used to construct B7-H3 overexpression or knockdown cell models, respectively. Cell counting kit-8 (CCK-8), 5-bromo-2'-deoxyuridine (BrdU), and scratch wound healing assays were employed for evaluating the effects of B7-H3 on CRC cell multiplication and migration. Besides, the regulatory relationship between miR-128 and B7-H3 was validated through dual-luciferase reporter gene assay, qRT-PCR, and western blotting. RESULTS: B7-H3 expression level was remarkably elevated in CRC tissues and cell lines, and its high expression level was associated with increased tumor size, positive lymph node metastasis, and increased T stage. In CRC cells, B7-H3 overexpression significantly facilitated the cell multiplication and migration, while B7-H3 knockdown worked oppositely. Moreover, B7-H3 was identified as a target of miR-128, and miR-128 negatively regulated B7-H3 expression in CRC cells. CONCLUSION: B7-H3 expression is upregulated in CRC tissues and cell lines, and B7-H3 participates in promoting the proliferation and migration of CRC cells. Besides, B7-H3 expression is negatively regulated by miR-128 in CRC.


Asunto(s)
Antígenos B7/metabolismo , Neoplasias Colorrectales/patología , MicroARNs/metabolismo , Antagomirs/metabolismo , Antígenos B7/antagonistas & inhibidores , Antígenos B7/genética , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/mortalidad , Femenino , Humanos , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Persona de Mediana Edad , Estadificación de Neoplasias , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Alineación de Secuencia , Tasa de Supervivencia
8.
Cancer Immunol Immunother ; 70(9): 2453-2465, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33543339

RESUMEN

BACKGROUND: T cell with chimeric antigen receptors (CAR-T) has presented remarkable efficacy for blood cancer as an emerging immunotherapy. However, for solid tumors, the therapeutic efficacy is much impaired due to the lack of infiltration and persistence of CAR-T in tumor tissue. Thus, we constructed an interleukin-7-loaded oncolytic adenovirus and combined the use of oncolytic virus and CAR-T to improve the therapeutic outcome. METHODS: We constructed an interleukin-7-loaded oncolytic adenovirus (oAD-IL7) and a B7H3-targeted CAR-T and explored the efficacy of the single use of oAD-IL7, B7H3-CAR-T, or the combined therapy for glioblastoma in vitro and in vivo. The improved CAR-T anti-tumor efficacy was evaluated according to the proliferation, survival, persistence, exhaustion of T cells, and tumor regression. RESULTS: Constructed oAD-IL7 and B7H3-CAR-T presented moderate cytotoxicity during in vitro study, but failed to induce a thorough and persistent anti-tumor therapeutic efficacy in vivo. The combination of oAD-IL7 and B7H3-CAR-T in vitro resulted in enhanced T cell proliferation and reduced T cell apoptosis. The joint efficacy was further confirmed using tumor-bearing xenograft mice. During in vivo study, the mice treated with both oAD-IL7 and B7H3-CAR-T showed prolonged survival and reduced tumor burden. According to the ex vivo study, oAD-IL7 improved the proliferation and persistence of tumor-infiltrating B7H3-CAR-T, but failed to reverse the exhaustion. CONCLUSIONS: Our results indicated that oAD-IL7 is a promising auxiliary therapy to improve the therapeutic efficacy of B7H3-CAR-T in glioblastoma by providing the activating signals for tumor-infiltrating T cells. Our results also lay the basis for the future clinical trials for the combination of IL7-loaded oncolytic adenovirus and CAR-T therapy for glioblastoma.


Asunto(s)
Terapia Genética , Vectores Genéticos/genética , Inmunoterapia Adoptiva , Interleucina-7/genética , Viroterapia Oncolítica , Virus Oncolíticos/genética , Adenoviridae/genética , Animales , Apoptosis/genética , Apoptosis/inmunología , Antígenos B7/antagonistas & inhibidores , Antígenos B7/inmunología , Antígenos B7/metabolismo , Citocinas/metabolismo , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Glioblastoma/etiología , Glioblastoma/metabolismo , Glioblastoma/patología , Glioblastoma/terapia , Humanos , Inmunofenotipificación , Inmunoterapia Adoptiva/métodos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Viroterapia Oncolítica/métodos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Clin Cancer Res ; 27(10): 2938-2946, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33619171

RESUMEN

PURPOSE: Patients with relapsed pediatric solid malignancies have few therapeutic options, and many of these patients die of their disease. B7-H3 is an immune checkpoint protein encoded by the CD276 gene that is overexpressed in many pediatric cancers. Here, we investigate the activity of the B7-H3-targeting antibody-drug conjugate (ADC) m276-SL-PBD in pediatric solid malignancy patient-derived (PDX) and cell line-derived xenograft (CDX) models. EXPERIMENTAL DESIGN: B7-H3 expression was quantified by RNA sequencing and by IHC on pediatric PDX microarrays. We tested the safety and efficacy of m276-SL-PBD in two stages. Randomized trials of m276-SL-PBD of 0.5 mg/kg on days 1, 8, and 15 compared with vehicle were performed in PDX or CDX models of Ewing sarcoma (N = 3), rhabdomyosarcoma (N = 4), Wilms tumors (N = 2), osteosarcoma (N = 5), and neuroblastoma (N = 12). We then performed a single mouse trial in 47 PDX or CDX models using a single 0.5 m/kg dose of m276-SL-PBD. RESULTS: The vast majority of PDX and CDX samples studied showed intense membranous B7-H3 expression (median H-score 177, SD 52). In the randomized trials, m276-SL-PBD showed a 92.3% response rate, with 61.5% of models showing a maintained complete response (MCR). These data were confirmed in the single mouse trial with an overall response rate of 91.5% and MCR rate of 64.4%. Treatment-related mortality rate was 5.5% with late weight loss observed in a subset of models dosed once a week for 3 weeks. CONCLUSIONS: m276-SL-PBD has significant antitumor activity across a broad panel of pediatric solid tumor PDX models.


Asunto(s)
Antígenos B7/antagonistas & inhibidores , Inmunoconjugados/farmacología , Neoplasias/tratamiento farmacológico , Animales , Antígenos B7/genética , Línea Celular Tumoral , Niño , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoconjugados/uso terapéutico , Ratones , Neoplasias/diagnóstico , Neoplasias/etiología , Neoplasias/metabolismo , Pediatría , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Naunyn Schmiedebergs Arch Pharmacol ; 394(6): 1057-1065, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33616679

RESUMEN

V-domain Ig suppressor of T cell activation (VISTA) is a transmembrane protein that plays a crucial role in the regulation of antitumor immunity. Therapeutic intervention that inhibits the VISTA pathway constitutes a new approach in the treatment of cancer. The aim of the present study was to provide a bibliometric literature review of VISTA research in the field of cancer. Published articles on the topic were retrieved from the Web of Science Core Collection Database of Hassan II University from the beginning of the database to November 14, 2019. The articles were analyzed and a lot of information was available on the Web of Science, such as the number of citations, the names of the authors, country, publication year, Web of Science categories, and journal. A total of 76 papers (research and review articles) were retrieved from the Web of Science Core Collection Database to introduce VISTA research in cancer topic. All of the articles were published in English during the period between 2011 and 2019; the annual publications number has increased from 1 in 2011 to 22 in 2019. Cancer immunology immunotherapy journal, Cancer immunology research journal, and Cancer research journal, each one has published 3 articles (3.9% of the total publications), the impact factors of the journals ranged from 2.34 to 10.19. The author who has published high number of articles was Noelle RJ with 11 articles; according to the keyword co-occurrence, VISTA was the most frequent keyword with a frequency of 42.1%, followed by immune (36.8%). This is the first work that treats the application of bibliometric methods in VISTA research in the oncology field and represents an important bibliographic source for future studies on the role of VISTA in cancer and immunotherapy of cancer.


Asunto(s)
Antígenos B7/antagonistas & inhibidores , Inmunoterapia/métodos , Neoplasias/terapia , Animales , Bibliometría , Humanos , Factor de Impacto de la Revista , Ratones , Neoplasias/inmunología , Publicaciones Periódicas como Asunto/estadística & datos numéricos
11.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33483421

RESUMEN

MYC is a powerful transcription factor overexpressed in many human cancers including B cell and prostate cancers. Antibody therapeutics are exciting opportunities to attack cancers but require knowledge of surface proteins that change due to oncogene expression. To identify how MYC overexpression remodels the cell surface proteome in a cell autologous fashion and in different cell types, we investigated the impact of MYC overexpression on 800 surface proteins in three isogenic model cell lines either of B cell or prostate cell origin engineered to have high or low MYC levels. We found that MYC overexpression resulted in dramatic remodeling (both up- and down-regulation) of the cell surfaceome in a cell type-dependent fashion. We found systematic and large increases in distinct sets of >80 transporters including nucleoside transporters and nutrient transporters making cells more sensitive to toxic nucleoside analogs like cytarabine, commonly used for treating hematological cancers. Paradoxically, MYC overexpression also increased expression of surface proteins driving cell turnover such as TNFRSF10B, also known as death receptor 5, and immune cell attacking signals such as the natural killer cell activating ligand NCR3LG1, also known as B7-H6. We generated recombinant antibodies to these two targets and verified their up-regulation in MYC overexpression cell lines and showed they were sensitive to bispecific T cell engagers (BiTEs). Our studies demonstrate how MYC overexpression leads to dramatic bidirectional remodeling of the surfaceome in a cell type-dependent but functionally convergent fashion and identify surface targets or combinations thereof as possible candidates for cytotoxic metabolite or immunotherapy.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Linfocitos B/efectos de los fármacos , Antígenos B7/genética , Células Epiteliales/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Anticuerpos Biespecíficos/biosíntesis , Linfocitos B/inmunología , Linfocitos B/patología , Antígenos B7/antagonistas & inhibidores , Antígenos B7/inmunología , Ingeniería Celular/métodos , Línea Celular Tumoral , Citarabina/farmacología , Células Epiteliales/inmunología , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunosupresores/farmacología , Inmunoterapia/métodos , Masculino , Terapia Molecular Dirigida/métodos , Plásmidos/química , Plásmidos/metabolismo , Próstata/inmunología , Próstata/patología , Unión Proteica , Proteínas Proto-Oncogénicas c-myc/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/antagonistas & inhibidores , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Transfección
12.
Clin Cancer Res ; 27(5): 1227-1235, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33051306

RESUMEN

The recent impressive clinical responses to antibody-based immunotherapy have prompted the identification of clinically relevant tumor antigens that can serve as targets in solid tumors. Among them, B7-H3, a member of the B7 ligand family, represents an attractive target for antibody-based immunotherapy, it is overexpressed on differentiated malignant cells and cancer-initiating cells, with limited heterogeneity, and high frequency (60% of 25,000 tumor samples) in many different cancer types, but has a limited expression at low level in normal tissues. In nonmalignant tissues, B7-H3 has a predominantly inhibitory role in adaptive immunity, suppressing T-cell activation and proliferation. In malignant tissues, B7-H3 inhibits tumor antigen-specific immune responses, leading to a protumorigenic effect. B7-H3 also has nonimmunologic protumorigenic functions, such as promoting migration and invasion, angiogenesis, chemoresistance, and endothelial-to-mesenchymal transition, as well as affecting tumor cell metabolism. As a result, B7-H3 expression in tumors is associated with poor prognosis. Although experimental B7-H3 silencing reduces cancer cell malignant potential, there has been limited emphasis on the development of B7-H3-blocking antibodies, most likely because the B7-H3 receptor remains unknown. Instead, many antibody-based strategies utilizing distinct effector mechanisms to target B7-H3-expressing cancer cells have been developed. These strategies have demonstrated potent antitumor activity and acceptable safety profiles in preclinical models. Ongoing clinical trials are assessing their safety and efficacy in patients. Identification of the B7-H3 receptor will improve our understanding of its role in tumor immunity, and will suggest rational strategies to develop blocking antibodies, which may enhance the therapeutic efficacy of tumor immunity.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos B7/antagonistas & inhibidores , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Humanos , Terapia Molecular Dirigida , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología
13.
Biochem Pharmacol ; 183: 114298, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33153969

RESUMEN

B7-H3 is an immune checkpoint molecule from the B7 superfamily. It has been widely studied in tumor immune evasion in certain types of cancer. In our preliminary study, we found that B7-H3 is specifically enriched in tumor-associated macrophages (TAMs) in triple-negative breast cancer (TNBC) patients and strongly correlated with poor clinical prognosis. However, the role of B7-H3 in breast cancer remains elusive. Our current study aims to explore the potential of B7-H3 as a novel target in TNBC therapy. Here, we demonstrated that B7-H3 enriched on TAMs is tightly correlated with TNBC clinical progression. B7-H3high TAMs exhibit great pro-metastatic and immunosuppressive functions by intriguing extracellular matrix (ECM) reconstruction and tumor angiogenesis, therefore helping tumor cell dissemination and dampening T-cell infiltration in tumor microenvironment (TME). Importantly, targeting blockade of B7-H3 by anti-B7-H3 antibody improves the tumor vasculature disorder, thereby enhancing chemotherapy and PD-1 therapy efficacy. In conclusion, our study establishes the correlation between B7-H3high TAMs and TNBC progression for the first time. By exploring the possibility of targeting B7-H3 expressed in both tumor cells and TAMs, we suggest that B7-H3 could be a promising target in clinical TNBC treatment.


Asunto(s)
Antígenos B7/biosíntesis , Sistemas de Liberación de Medicamentos/métodos , Neovascularización Patológica/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Macrófagos Asociados a Tumores/metabolismo , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antígenos B7/antagonistas & inhibidores , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/biosíntesis , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Paclitaxel/administración & dosificación , Células RAW 264.7 , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/fisiología , Macrófagos Asociados a Tumores/efectos de los fármacos , Macrófagos Asociados a Tumores/patología
14.
J Cancer Res Clin Oncol ; 147(2): 517-531, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33237432

RESUMEN

OBJECTIVE: V-domain Ig suppressor of T cell activation (VISTA) is a novel immune checkpoint protein that belongs to the B7 family. The aim of this study was to investigate the prognostic significance and therapeutic potential of VISTA in patients with pancreatic cancer. METHODS: Using immunohistochemistry (IHC), we examined the expression of VISTA and demonstrated the associations between the VISTA and overall survival in 223 PDAC patients from 2 different unrelated retrospective cohorts. The multiplex immunofluorescence was performed to illuminate the relationship between VISTA expression and tumor-infiltrating immune cell subclusters of PDAC. We also verified the findings in The Cancer Genome Atlas (TCGA) dataset. The anti-tumor effect of anti-VISTA therapy was studied by the mouse model with liver metastases of PDAC. RESULTS: The VISTA protein was highly expressed in 25.6% of tumor cells (TCs), 38.1% of immune cells, and 26.0% of endothelial cells in 223 PDAC tumor tissues. VISTA expression in TCs was significantly associated with prolonged overall survival. Multiplex immunofluorescence analysis revealed that VISTA level was positively correlated with CD68+ macrophages, CD3+ T cells, and CD19+ B cells in PDAC. However, a higher expression level of VISTA was detected in tumor-infiltrating CD68+ macrophages than in CD3+ T and CD19+ B cells. Furthermore, anti-VISTA antibody treatment significantly reduced the number of metastatic nodules in livers of mouse models of PDAC with liver metastases. CONCLUSION: VISTA expressed in TCs is associated with a favorable prognosis in PDAC. Moreover, immunotherapy with anti-VISTA antibodies may potentially be an effective treatment strategy against PDAC.


Asunto(s)
Antígenos B7/análisis , Carcinoma Ductal Pancreático/mortalidad , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Pancreáticas/mortalidad , Antígenos B7/antagonistas & inhibidores , Antígeno B7-H1/análisis , Carcinoma Ductal Pancreático/química , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/secundario , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/secundario , Neoplasias Pancreáticas/química , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Pronóstico , Microambiente Tumoral
15.
Sci Rep ; 10(1): 15171, 2020 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-32938950

RESUMEN

V-domain immunoglobulin (Ig) suppressor of T cell activation (VISTA) is an immune checkpoint that maintains peripheral T cell quiescence and inhibits anti-tumor immune responses. VISTA functions by dampening the interaction between myeloid cells and T cells, orthogonal to PD-1 and other checkpoints of the tumor-T cell signaling axis. Here, we report the use of yeast surface display to engineer an anti-VISTA antibody that binds with high affinity to mouse, human, and cynomolgus monkey VISTA. Our anti-VISTA antibody (SG7) inhibits VISTA function and blocks purported interactions with both PSGL-1 and VSIG3 proteins. SG7 binds a unique epitope on the surface of VISTA, which partially overlaps with other clinically relevant antibodies. As a monotherapy, and to a greater extent as a combination with anti-PD1, SG7 slows tumor growth in multiple syngeneic mouse models. SG7 is a promising clinical candidate that can be tested in fully immunocompetent mouse models and its binding epitope can be used for future campaigns to develop species cross-reactive inhibitors of VISTA.


Asunto(s)
Anticuerpos/metabolismo , Antígenos B7/antagonistas & inhibidores , Epítopos/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Animales , Reacciones Antígeno-Anticuerpo , Antígenos B7/genética , Antígenos B7/inmunología , Sitios de Unión , Moléculas de Adhesión Celular/metabolismo , Técnicas de Visualización de Superficie Celular , Reacciones Cruzadas , Epítopos/genética , Femenino , Humanos , Inmunoglobulinas/metabolismo , Macaca fascicularis , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Moleculares , Unión Proteica , Ingeniería de Proteínas
16.
Mol Cancer Ther ; 19(11): 2235-2244, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32967924

RESUMEN

B7-H3, also referred to as CD276, is a member of the B7 family of immune regulatory proteins. B7-H3 is overexpressed on many solid cancers, including prostate cancer, renal cell carcinoma, melanoma, squamous cell carcinoma of the head and neck, non-small cell lung cancer, and breast cancer. Overexpression of B7-H3 is associated with disease severity, risk of recurrence and reduced survival. In this article, we report the preclinical development of MGC018, an antibody-drug conjugate targeted against B7-H3. MGC018 is comprised of the cleavable linker-duocarmycin payload, valine-citrulline-seco duocarmycin hydroxybenzamide azaindole (vc-seco-DUBA), conjugated to an anti-B7-H3 humanized IgG1/kappa mAb through reduced interchain disulfides, with an average drug-to-antibody ratio of approximately 2.7. MGC018 exhibited cytotoxicity toward B7-H3-positive human tumor cell lines, and exhibited bystander killing of target-negative tumor cells when cocultured with B7-H3-positive tumor cells. MGC018 displayed potent antitumor activity in preclinical tumor models of breast, ovarian, and lung cancer, as well as melanoma. In addition, antitumor activity was observed toward patient-derived xenograft models of breast, prostate, and head and neck cancer displaying heterogeneous expression of B7-H3. Importantly, MGC018 exhibited a favorable pharmacokinetic and safety profile in cynomolgus monkeys following repeat-dose administration. The antitumor activity observed preclinically with MGC018, together with the positive safety profile, provides evidence of a potentially favorable therapeutic index and supports the continued development of MGC018 for the treatment of solid cancers. GRAPHICAL ABSTRACT: http://mct.aacrjournals.org/content/molcanther/19/11/2235/F1.large.jpg.


Asunto(s)
Antígenos B7/antagonistas & inhibidores , Evaluación Preclínica de Medicamentos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoconjugados/farmacología , Neoplasias/tratamiento farmacológico , Animales , Antígenos B7/genética , Antígenos B7/metabolismo , Efecto Espectador , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Monitoreo de Drogas , Técnicas de Silenciamiento del Gen , Humanos , Inhibidores de Puntos de Control Inmunológico/química , Inhibidores de Puntos de Control Inmunológico/aislamiento & purificación , Inmunoconjugados/química , Inmunoconjugados/aislamiento & purificación , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Theranostics ; 10(23): 10498-10512, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32929362

RESUMEN

Rationale: Current traditional treatment options are frequently ineffective to fight against ovarian cancer due to late diagnosis and high recurrence. Therefore, there is a vital need for the development of novel therapeutic agents. B7H3, an immune checkpoint protein, is highly expressed in various cancers, representing it a promising target for cancer immunotherapy. Although targeting B7H3 by bispecific T cell-engaging antibodies (BiTE) has achieved successes in hematological malignancies during recent years, attempts to use them for the treatment of solid cancers are less favorable, in part due to the heterogeneity of tumors. Sorafenib is an unselective inhibitor of multiple kinases currently being tested in clinical trials for several tumors, including ovarian cancer which showed limited activity and inevitable side effect for ovarian cancer treatment. However, it is able to enhance antitumor immune response, which indicates sorafenib may improve the efficiency of immunotherapy. Methods: We evaluated the expression of B7H3 in ovarian cancer using online database and validated its expression of tumor tissues by immunohistochemistry staining. Then, B7H3 expression and the effects of sorafenib on ovarian cancer cell lines were determined by flow cytometry. In addition, 2D and 3D ovarian cancer models were established to test the combined therapeutic effect in vitro. Finally, the efficiency of B7H3×CD3 BiTE alone and its combination with sorafenib were evaluated both in vitro and in vivo. Results: Our data showed that B7H3 was highly expressed in ovarian cancer compared with normal samples. Treatment with sorafenib inhibited ovarian cancer cell proliferation and induced a noticeable upregulation of B7H3 expression level. Further study suggested that B7H3×CD3 BiTE was effective in mediating T cell killing to cancer cells. Combined treatment of sorafenib and B7H3×CD3 BiTE had synergistic anti-tumor effects in ovarian cancer models. Conclusions: Overall, our study indicates that combination therapy with sorafenib and B7H3×CD3 BiTE may be a new therapeutic option for the further study of preclinical treatment of OC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Antígenos B7/antagonistas & inhibidores , Carcinoma Epitelial de Ovario/terapia , Neoplasias Ováricas/terapia , Sorafenib/farmacología , Animales , Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Antígenos B7/análisis , Antígenos B7/metabolismo , Complejo CD3/antagonistas & inhibidores , Carcinoma Epitelial de Ovario/inmunología , Carcinoma Epitelial de Ovario/mortalidad , Carcinoma Epitelial de Ovario/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Conjuntos de Datos como Asunto , Sinergismo Farmacológico , Femenino , Células HEK293 , Humanos , Estimación de Kaplan-Meier , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Recurrencia Local de Neoplasia , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Ovario/patología , Sorafenib/uso terapéutico , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Am Chem Soc ; 142(38): 16194-16198, 2020 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-32894020

RESUMEN

V-domain Ig suppressor of T-cell activation (VISTA) is an immune checkpoint that affects the ability of T-cells to attack tumors. A FRET-based high throughput screening identified NSC622608 as the first small-molecule ligand for VISTA. Investigation of the interaction of NSC622608 with VISTA using STD NMR and molecular modeling enabled the identification of a potential binding site in VISTA for NSC622608. Screening NSC622608 against a library of single-point VISTA mutants revealed the key residues in VISTA interacting with NSC622608. Further structural optimization resulted in a lead with submicromolar VISTA binding affinity. The lead compound blocked VISTA signaling in vitro, enhanced T-cell proliferation, and restored T-cell activation in the presence of VISTA-expressing cancer cell lines. This work would enable future development of small molecules targeting VISTA as immunomodulators and imaging probes.


Asunto(s)
Antígenos B7/antagonistas & inhibidores , Descubrimiento de Drogas , Inhibidores de Puntos de Control Inmunológico/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Antígenos B7/inmunología , Línea Celular , Humanos , Inhibidores de Puntos de Control Inmunológico/química , Ligandos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/química , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
19.
Biosci Rep ; 40(7)2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32602545

RESUMEN

Immune checkpoint blockade treatments bring remarkable clinical benefits to fighting several solid malignancies. However, the efficacy of immune checkpoint blockade in breast cancer remains controversial. Several clinical trials of immune checkpoint blockades focused on the effect of CTLA4 and PD1/PDL1 checkpoint inhibitors on breast cancer. Only a small portion of patients benefited from these therapies. Here we systematically investigated the expression of 50 immune checkpoint genes, including ADORA2A, LAG-3, TIM-3, PD1, PDL1, PDL2, CTLA-4, IDO1, B7-H3, B7-H4, CD244, BTLA, TIGIT, CD80, CD86, VISTA, CD28, ICOS, ICOSLG, HVEM, CD160, LIGHT, CD137, CD137L, OX40, CD70, CD27, CD40, CD40LG, LGALS9, GITRL, CEACAM1, CD47, SIRPA, DNAM1, CD155, 2B4, CD48, TMIGD2, HHLA2, BTN2A1, DC-SIGN, BTN2A2, BTN3A1, BTNL3, BTNL9, CD96, TDO, CD200 and CD200R, in different subtypes of breast cancer and assessed their prognostic value. The results showed that the expression patterns of these 50 immune checkpoint genes were distinct in breast cancer. High expression of B7-H3 mRNA was significantly associated with worse overall survival (OS), especially in patients with luminal A and luminal B breast cancer. The mRNA expression levels of TIM-3, ADORA2A, LAG3, CD86, CD80, PD1 and IDO1 had no relationship with OS in breast cancer. High expression levels of CTLA-4 and TIGIT were correlated with favorable prognosis in breast cancer. Interestingly, we observed that B7-H3 expression was negatively correlated with the efficacy of cyclophosphamide (CTX). In summary, our study suggested that B7-H3 has potential prognostic value in breast cancer and is a promising target for immune therapy.


Asunto(s)
Antineoplásicos/farmacología , Antígenos B7/metabolismo , Neoplasias de la Mama/mortalidad , Proteínas de Punto de Control Inmunitario/metabolismo , Antineoplásicos/uso terapéutico , Antígenos B7/análisis , Antígenos B7/antagonistas & inhibidores , Mama/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Ciclofosfamida/farmacología , Ciclofosfamida/uso terapéutico , Resistencia a Antineoplásicos/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Proteínas de Punto de Control Inmunitario/análisis , Estimación de Kaplan-Meier , Pronóstico
20.
J Hematol Oncol ; 13(1): 83, 2020 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-32600443

RESUMEN

VISTA (V-domain immunoglobulin suppressor of T cell activation) is a well-established immune regulatory receptor. However, pre-clinical investigations indicated more complicated influences of VISTA on cancer immunity than previously recognized. Here, we review the current knowledge on the therapeutic phenotypes and molecular mechanisms that underlie the contradictory roles of VISTA in checking anti-cancer immune responses. Furthermore, we highlight the potential indeterminacy of VISTA-targeted strategies in cancer immunotherapy, with in silico analyses. In fact, VISTA functions like a homeostatic regulator that actively normalizes immune responses. Thus, the regulatory role of VISTA in anti-cancer immunity remains to be fully elucidated.


Asunto(s)
Antígenos B7/antagonistas & inhibidores , Proteínas de Punto de Control Inmunitario/fisiología , Inmunoterapia , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/terapia , Subgrupos de Linfocitos T/inmunología , Antígenos B7/química , Antígenos B7/inmunología , Ensayos Clínicos como Asunto , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Proteínas de Punto de Control Inmunitario/química , Proteínas de Punto de Control Inmunitario/inmunología , Inmunomodulación , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Proteínas de Neoplasias/química , Proteínas de Neoplasias/inmunología , Neoplasias/inmunología , Especificidad de Órganos , Pronóstico , Escape del Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA