Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cancer Sci ; 115(4): 1114-1128, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38332689

RESUMEN

The direction and magnitude of immune responses are critically affected when dead cells are disposed of. Milk fat globule-epidermal growth factor-factor 8 (MFG-E8) promotes the engulfment of apoptotic normal and cancerous cells without inducing inflammation. We have previously reported that a certain proportion of the cancer cells express abundant MFG-E8, and that such expression is associated with the shorter survival of patients with esophageal cancer who had received chemotherapy before surgery. However, the influence of tumor-derived and systemically existing MFG-E8 on antitumor immune responses has not yet been fully investigated. Herein, we showed that CTL-dependent antitumor immune responses were observed in mice with no or decreased levels of systemic MFG-E8, and that such responses were enhanced further with the administration of anti-PD-1 antibody. In mice with decreased levels of systemic MFG-E8, the dominance of regulatory T cells in tumor-infiltrating lymphocytes was inverted to CD8+ T cell dominance. MFG-E8 expression by tumor cells appears to affect antitumor immune responses only when the level of systemic MFG-E8 is lower than the physiological status. We have also demonstrated in the clinical setting that lower levels of plasma MFG-E8, but not MFG-E8 expression in tumor cells, before the treatment was associated with objective responses to anti-PD-1 therapy in patients with non-small cell lung cancer. These results suggest that systemic MFG-E8 plays a critical role during the immunological initiation process of antigen-presenting cells to increase tumor-specific CTLs. Regulation of the systemic level of MFG-E8 might induce efficient antitumor immune responses and enhance the potency of anti-PD-1 therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Esofágicas , Neoplasias Pulmonares , Animales , Humanos , Ratones , Antígenos de Superficie/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Esofágicas/tratamiento farmacológico , Inflamación/patología , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas de la Leche/metabolismo , Linfocitos T Citotóxicos/metabolismo
2.
Int J Clin Oncol ; 29(6): 771-779, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38600426

RESUMEN

BACKGROUND: Adenocarcinomas show a stepwise progression from atypical adenomatous hyperplasia (AAH) through adenocarcinoma in situ (AIS) to invasive adenocarcinoma (IA). Immunoglobulin superfamily containing leucine-rich repeat (ISLR) is a marker of tumor-restraining cancer-associated fibroblasts (CAFs), which are distinct from conventional, strongly α-smooth muscle actin (αSMA)-positive CAFs. Fibroblast activation protein (FAP) has been focused on as a potential therapeutic and diagnostic target of CAFs. METHODS: We investigated the changes in protein expression during adenocarcinoma progression in the pre-existing alveolar septa by assessing ISLR, αSMA, and FAP expression in normal lung, AAH, AIS, and IA. Fourteen AAH, seventeen AIS, and twenty IA lesions were identified and randomly sampled. Immunohistochemical analysis was performed to evaluate cancer-associated changes and FAP expression in the pre-existing alveolar structures. RESULTS: Normal alveolar septa expressed ISLR. The ISLR level in the alveolar septa decreased in AAH and AIS tissues when compared with that in normal lung tissue. The αSMA-positive area gradually increased from the adjacent lung tissue (13.3% ± 15%) to AIS (87.7% ± 14%), through AAH (70.2% ± 21%). Moreover, the FAP-positive area gradually increased from AAH (1.69% ± 1.4%) to IA (11.8% ± 7.1%), through AIS (6.11% ± 5.3%). Protein expression changes are a feature of CAFs in the pre-existing alveolar septa that begin in AAH. These changes gradually progressed from AAH to IA through AIS. CONCLUSIONS: FAP-positive fibroblasts may contribute to tumor stroma formation in early-stage lung adenocarcinoma, and this could influence the development of therapeutic strategies targeting FAP-positive CAFs for disrupting extracellular matrix formation.


Asunto(s)
Adenocarcinoma del Pulmón , Progresión de la Enfermedad , Endopeptidasas , Neoplasias Pulmonares , Proteínas de la Membrana , Humanos , Adenocarcinoma del Pulmón/patología , Adenocarcinoma del Pulmón/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Masculino , Femenino , Persona de Mediana Edad , Proteínas de la Membrana/metabolismo , Anciano , Gelatinasas/metabolismo , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/genética , Actinas/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Biomarcadores de Tumor/metabolismo , Alveolos Pulmonares/patología , Alveolos Pulmonares/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Estadificación de Neoplasias , Adenocarcinoma in Situ/patología , Adenocarcinoma in Situ/metabolismo , Adulto
3.
Int Immunol ; 34(5): 277-289, 2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-35094065

RESUMEN

Effective tumor immunotherapy requires physical contact of T cells with cancer cells. However, tumors often constitute a specialized microenvironment that excludes T cells from the vicinity of cancer cells, and its underlying mechanisms are still poorly understood. DOCK2 is a Rac activator critical for migration and activation of lymphocytes. We herein show that cancer-derived cholesterol sulfate (CS), a lipid product of the sulfotransferase SULT2B1b, acts as a DOCK2 inhibitor and prevents tumor infiltration by effector T cells. Using clinical samples, we found that CS was abundantly produced in certain types of human cancers such as colon cancers. Functionally, CS-producing cancer cells exhibited resistance to cancer-specific T-cell transfer and immune checkpoint blockade. Although SULT2B1b is known to sulfate oxysterols and inactivate their tumor-promoting activity, the expression levels of cholesterol hydroxylases, which mediate oxysterol production, are low in SULT2B1b-expressing cancers. Therefore, SULT2B1b inhibition could be a therapeutic strategy to disrupt tumor immune evasion in oxysterol-non-producing cancers. Thus, our findings define a previously unknown mechanism for tumor immune evasion and provide a novel insight into the development of effective immunotherapies.


Asunto(s)
Neoplasias , Oxiesteroles , Ésteres del Colesterol/metabolismo , Humanos , Inmunoterapia , Linfocitos T/metabolismo , Microambiente Tumoral
4.
J Virol ; 95(9)2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33627393

RESUMEN

Herpes simplex virus (HSV) is a promising tool for developing oncolytic virotherapy. We recently reported a platform for receptor-retargeted oncolytic HSVs that incorporates single-chain antibodies (scFvs) into envelope glycoprotein D (gD) to mediate virus entry via tumor-associated antigens. Therefore, it would be useful to develop an efficient system that can screen antibodies that might mediate HSV entry when they are incorporated as scFvs into gD. We created an HSV-based screening probe by the genetic fusion of a gD mutant with ablated binding capability to the authentic HSV entry receptors and the antibody-binding C domain of streptococcal protein G. This engineered virus failed to enter cells through authentic receptors. In contrast, when this virus was conjugated with an antibody specific to an antigen on the cell membrane, it specifically entered cells expressing the cognate antigen. This virus was used as a probe to identify antibodies that mediate virus entry via recognition of certain molecules on the cell membrane other than authentic receptors. Using this method, we identified an antibody specific to epiregulin (EREG), which has been investigated mainly as a secreted growth factor and not necessarily for its precursor that is expressed in a transmembrane form. We constructed an scFv from the anti-EREG antibody for insertion into the retargeted HSV platform and found that the recombinant virus entered cells specifically through EREG expressed by the cells. This novel antibody-screening system may contribute to the discovery of unique and unexpected molecules that might be used for the entry of receptor-retargeted oncolytic HSVs.IMPORTANCE The tropism of the cellular entry of HSV is dependent on the binding of the envelope gD to one of its authentic receptors. This can be fully retargeted to other receptors by inserting scFvs into gD with appropriate modifications. In theory, upon binding to the engineered gD, receptors other than authentic receptors should induce a conformational change in the gD, which activates downstream mechanisms required for viral entry. However, prerequisite factors for receptors to be used as targets of a retargeted virus remain poorly understood, and it is difficult to predict which molecules might be suitable for our retargeted HSV construct. Our HSV-based probe will allow unbiased screening of antibody-antigen pairs that mediate virus entry and might be a useful tool to identify suitable pairs for our construct and to enhance our understanding of virus-cell interactions during infection by HSV and possibly other viruses.


Asunto(s)
Epirregulina/metabolismo , Herpesvirus Humano 1/metabolismo , Virus Oncolíticos/fisiología , Anticuerpos de Cadena Única/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Animales , Células CHO , Línea Celular Tumoral , Chlorocebus aethiops , Cricetulus , Humanos , Neoplasias/terapia , Viroterapia Oncolítica , Células Vero , Tropismo Viral
5.
Cancer Sci ; 112(9): 3484-3490, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34187084

RESUMEN

For successful immunotherapy for cancer, it is important to understand the immunological status of tumor antigen-specific CD8+ T cells in the tumor microenvironment during tumor progression. In this study, we monitored the behavior of B16OVA-Luc cells in mice immunized with a model tumor antigen ovalbumin (OVA). Using bioluminescence imaging, we identified the time series of OVA-specific CD8+ T-cell responses during tumor progression: initial progression, immune control, and the escape phase. As a result of analyzing the status of tumor antigen-specific CD8+ cells in those 3 different phases, we found that the expression of NKG2D defines tumor-reacting effector CD8+ T cells. NKG2D may control the fate and TOX expression of tumor-reacting CD8+ T cells, considering that NKG2D blockade in OVA-vaccinated mice delayed the growth of the B16OVA-Luc2 tumor and increased the presence of tumor-infiltrating OVA-specific CD8+ T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Microambiente Tumoral/inmunología , Animales , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/metabolismo , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/farmacocinética , Bromodesoxiuridina/administración & dosificación , Bromodesoxiuridina/farmacocinética , Interferón gamma/deficiencia , Interferón gamma/genética , Luciferasas/metabolismo , Mediciones Luminiscentes/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/administración & dosificación , Ovalbúmina/metabolismo , Neoplasias Cutáneas/patología , Vacunación/métodos
6.
Cancer Sci ; 112(4): 1633-1643, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33565179

RESUMEN

Tumor metastasis is the leading cause of death worldwide and involves an extremely complex process composed of multiple steps. Our previous study demonstrated that apoptosis signal-regulating kinase 1 (ASK1) deficiency in mice attenuates tumor metastasis in an experimental lung metastasis model. However, the steps of tumor metastasis regulated by ASK1 remain unclear. Here, we showed that ASK1 deficiency in mice promotes natural killer (NK) cell-mediated intravascular tumor cell clearance in the initial hours of metastasis. In response to tumor inoculation, ASK1 deficiency upregulated immune response-related genes, including interferon-gamma (IFNγ). We also revealed that NK cells are required for these anti-metastatic phenotypes. ASK1 deficiency augmented cytokine production chemoattractive to NK cells possibly through induction of the ligand for NKG2D, a key activating receptor of NK cells, leading to further recruitment of NK cells into the lung. These results indicate that ASK1 negatively regulates NK cell-dependent anti-tumor immunity and that ASK1-targeted therapy can provide a new tool for cancer immunotherapy to overcome tumor metastasis.


Asunto(s)
Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , MAP Quinasa Quinasa Quinasa 5/metabolismo , Metástasis de la Neoplasia/patología , Animales , Línea Celular , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Inmunoterapia/métodos , Interferón gamma/metabolismo , Células Asesinas Naturales/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/inmunología , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/inmunología , Células RAW 264.7
7.
Cancer Immunol Immunother ; 70(9): 2529-2543, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33570675

RESUMEN

Intravesical Bovis bacillus Calmette-Guérin (BCG) therapy is the most effective immunotherapy for bladder cancer, but it sometime causes serious side effects because of its inclusion of live bacteria. It is necessary to develop a more active but less toxic immunotherapeutic agent. Trehalose 6,6'-dimycolate (TDM), the most abundant hydrophobic glycolipid of the BCG cell wall, has been reported to show various immunostimulatory activities such as granulomagenesis and adjuvant activity. Here, we developed cationic liposomes incorporating TDM purified from Mycobacterium bovis BCG Connaught, and we investigated the antitumor effect of the cationic liposome TDM (Lip-TDM). Lip-TDM exerted an antitumor effect in bladder cancer, colon cancer, and melanoma-bearing mouse models that was comparable or even superior to that of BCG, with no body weight loss or granuloma formation. The antitumor effect of Lip-TDM disappeared in two types of mice: those with depletion of CD8+ T cells, and those with knockout of macrophage-inducible C-type lectin (Mincle) which recognize TDM. Lip-TDM treatment enhanced the maturation and migration of dendritic cells in the tumor microenvironment in a Mincle-dependent manner. Our results elucidate mechanisms that underlie Lip-TDM treatment and suggest that Lip-TDM has potential as a safe and effective treatment for various cancers.


Asunto(s)
Antineoplásicos Inmunológicos/administración & dosificación , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Factores Cordón/administración & dosificación , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Factores Inmunológicos/administración & dosificación , Mycobacterium bovis , Adyuvantes Inmunológicos , Animales , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/aislamiento & purificación , Linfocitos T CD8-positivos/metabolismo , Fraccionamiento Químico , Factores Cordón/química , Factores Cordón/aislamiento & purificación , Citocinas/metabolismo , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Factores Inmunológicos/química , Factores Inmunológicos/aislamiento & purificación , Inmunofenotipificación , Infusiones Parenterales , Liposomas , Activación de Linfocitos , Ratones , Estructura Molecular , Mycobacterium bovis/química , Solventes , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Sci ; 111(8): 2770-2778, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32573072

RESUMEN

Thalidomide and its analogues are known as immunomodulatory drugs (IMiDs) that possess direct antimyeloma effects, in addition to other secondary effects, including antiangiogenic, antiinflammatory, and immunomodulatory effects. Although the involvement of natural killer (NK) cells in the antitumor effects of IMiDs has been reported, it is unclear whether IMiDs inhibit cancer cell metastasis by regulating the antitumor function of NK cells. In this study, we examined the protective effects of thalidomide against cancer metastasis by focusing on its immunomodulatory effects through NK cells. Using experimental lung metastasis models, we found that pharmacological effects of thalidomide on host cells, but not its direct anticancer tumor effects, are responsible for the inhibition of lung metastases. To exert the antimetastatic effects of thalidomide, both γ-interferon (IFN-γ) production and direct cytotoxicity of NK cells were essential, without notable contribution from T cells. In thalidomide-treated mice, there was a significant increase in the terminally differentiated mature CD27lo NK cells in the peripheral tissues and NK cells in thalidomide-treated mice showed significantly higher cytotoxicity and IFN-γ production. The NK cell expression of T-bet was upregulated by thalidomide treatment and the downregulation of glycogen synthase kinase-3ß expression was observed in thalidomide-treated NK cells. Collectively, our study suggests that thalidomide induces the functional maturation of peripheral NK cells through alteration of T-bet expression to inhibit lung metastasis of cancer cells.


Asunto(s)
Citotoxicidad Inmunológica/efectos de los fármacos , Factores Inmunológicos/farmacología , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/tratamiento farmacológico , Talidomida/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , Ensayos de Selección de Medicamentos Antitumorales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/inmunología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Factores Inmunológicos/uso terapéutico , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Ratones , Ratones Noqueados , Proteínas de Dominio T Box/metabolismo , Talidomida/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
9.
Cancer Sci ; 109(9): 2670-2676, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29927042

RESUMEN

Accumulating evidence indicates the importance of natural killer (NK) cells in controlling tumor growth and metastasis. NK cell subsets display diversities in their function and tissue distribution and Mac-1hi CD27lo NK cells are the predominant population of lung-resident NK cells. Although the lung is a major organ where primary tumor develops and cancer cells metastasize, there is no clear evidence whether circulating NK cells and/or tissue-resident NK cells control tumor growth in the lung. In the present study, we examined an antitumor function of lung-resident NK cells to control pulmonary tumor growth. In an orthotopic lung tumor model, NK cells controlled pulmonary tumor growth, and mature circulating NK cell subsets were increased in tumor-bearing lungs through a C-X-C motif chemokine receptor 3 (CXCR3)-dependent mechanism. Although such increase in migratory NK cell subsets can be blocked by anti-CXCR3 treatment, there was no difference in pulmonary tumor growth in anti-CXCR3-treated mice compared with control mice. In addition to pulmonary tumor growth, lung-resident NK cells, but not migratory NK cells, play a dominant role in controlling metastatic growth of cancer cells in lung. These results strongly indicate an importance of lung-resident NK cells for controlling pulmonary tumor growth.


Asunto(s)
Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/patología , Pulmón/inmunología , Animales , Proliferación Celular , Neoplasias Pulmonares/inmunología , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Receptores CXCR3/antagonistas & inhibidores
10.
Cancer Sci ; 109(11): 3393-3402, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30156356

RESUMEN

Milk fat globule-epidermal growth factor factor 8 (MFG-E8) is secreted from macrophages and is known to induce immunological tolerance mediated by regulatory T cells. However, the roles of the MFG-E8 that is expressed by cancer cells have not yet been fully examined. Expression of MFG-E8 was examined using immunohistochemistry in surgical samples from 134 patients with esophageal squamous cell carcinoma. The relationships between MFG-E8 expression levels and clinicopathological factors, including tumor-infiltrating lymphocytes, were evaluated. High MFG-E8 expression was observed in 23.9% of the patients. The patients with tumors highly expressing MFG-E8 had a significantly higher percentage of neoadjuvant chemotherapy (NAC) history (P < .0001) and shorter relapse-free survival (P = 0.012) and overall survival (OS; P = .0047). On subgroup analysis, according to NAC history, patients with high MFG-E8 expression had significantly shorter relapse-free survival (P = .027) and OS (P = .0039) only when they had been treated with NAC. Furthermore, tumors with high MFG-E8 expression had a significantly lower ratio of CD8+ T cells/regulatory T cells in tumor-infiltrating lymphocytes (P = .042) only in the patients treated with NAC, and those with a lower ratio had a shorter OS (P = .026). High MFG-E8 expression was also found to be an independent prognostic factor in multivariate analysis. The abundant MFG-E8 expression in esophageal squamous cell carcinoma might have a negative influence on the long-term survival of patients after chemotherapy by affecting T-cell regulation in the tumor microenvironment.


Asunto(s)
Antígenos de Superficie/metabolismo , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Quimioterapia/métodos , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Proteínas de la Leche/metabolismo , Regulación hacia Arriba , Animales , Linfocitos T CD8-positivos , Carcinoma de Células Escamosas/cirugía , Supervivencia sin Enfermedad , Neoplasias Esofágicas/cirugía , Carcinoma de Células Escamosas de Esófago , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Terapia Neoadyuvante , Pronóstico , Resultado del Tratamiento , Microambiente Tumoral
11.
Cancer Sci ; 108(5): 1022-1031, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28266140

RESUMEN

Promising antitumor activities of nivolumab, a fully humanized IgG4 inhibitor antibody against the programmed death-1 protein, were suggested in previous phase 1 studies. The present phase 2, single-arm study (JAPIC-CTI #111681) evaluated the antitumor activities of nivolumab and explored its predictive correlates in advanced melanoma patients at 11 sites in Japan. Intravenous nivolumab 2 mg/kg was given repeatedly at 3-week intervals to 35 of 37 patients enrolled from December 2011 to May 2012 until they experienced unacceptable toxicity, disease progression, or complete response. Primary endpoint was objective response rate. Serum levels of immune modulators were assessed at multiple time points. As of 21 October 2014, median response duration, median progression-free survival, and median overall survival were 463 days, 169 days, and 18.0 months, respectively. The overall response rate and 1- and 2-year survival rates were 28.6%, 54.3%, and 42.9%, respectively. Thirteen patients remained alive at the end of the observation period and no deaths were drug related. Grade 3-4 drug-related adverse events were observed in 31.4% of patients. Pretreatment serum interferon-γ, and interleukin-6 and -10 levels were significantly higher in the patients with objective tumor responses than in those with tumor progression. In conclusion, giving repeated i.v. nivolumab had potent and durable antitumor effects and a manageable safety profile in advanced melanoma patients, strongly suggesting the usefulness of nivolumab for advanced melanoma and the usefulness of pretreatment serum cytokine profiles as correlates for predicting treatment efficacy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Biomarcadores/metabolismo , Citocinas/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Anciano , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Humanos , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Masculino , Melanoma/patología , Nivolumab , Tasa de Supervivencia
12.
J Virol ; 90(24): 11096-11105, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27707922

RESUMEN

Membrane fusion, which is the key process for both initial cell entry and subsequent lateral spread of herpes simplex virus (HSV), requires the four envelope glycoproteins gB, gD, gH, and gL. Syncytial mutations, predominantly mapped to the gB and gK genes, confer hyperfusogenicity on HSV and cause multinucleated giant cells, termed syncytia. Here we asked whether interaction of gD with a cognate entry receptor remains indispensable for initiating membrane fusion of syncytial strains. To address this question, we took advantage of mutant viruses whose viral entry into cells relies on the uniquely specific interaction of an engineered gD with epidermal growth factor receptor (EGFR). We introduced selected syncytial mutations into gB and/or gK of the EGFR-retargeted HSV and found that these mutations, especially when combined, enabled formation of extensive syncytia by human cancer cell lines that express the target receptor; these syncytia were substantially larger than the plaques formed by the parental retargeted HSV strain. We assessed the EGFR dependence of entry and spread separately by using direct entry and infectious center assays, respectively, and we found that the syncytial mutations did not override the receptor specificity of the retargeted viruses at either stage. We discuss the implications of these results for the development of more effective targeted oncolytic HSV vectors. IMPORTANCE: Herpes simplex virus (HSV) is investigated not only as a human pathogen but also as a promising agent for oncolytic virotherapy. We previously showed that both the initial entry and subsequent lateral spread of HSV can be retargeted to cells expressing tumor-associated antigens by single-chain antibodies fused to a receptor-binding-deficient envelope glycoprotein D (gD). Here we introduced syncytial mutations into the gB and/or gK gene of gD-retargeted HSVs to determine whether viral tropism remained dependent on the interaction of gD with the target receptor. Entry and spread profiles of the recombinant viruses indicated that gD retargeting does not abolish the hyperfusogenic activity of syncytial mutations and that these mutations do not eliminate the dependence of HSV entry and spread on a specific gD-receptor interaction. These observations suggest that syncytial mutations may be valuable for increasing the tumor-specific spreading of retargeted oncolytic HSV vectors.


Asunto(s)
Receptores ErbB/metabolismo , Herpesvirus Humano 1/genética , Mutación , Receptores Virales/metabolismo , Proteínas del Envoltorio Viral/genética , Animales , Células CHO , Línea Celular Tumoral , Supervivencia Celular , Chlorocebus aethiops , Cricetulus , Receptores ErbB/genética , Expresión Génica , Células Gigantes/metabolismo , Células Gigantes/ultraestructura , Células Gigantes/virología , Herpesvirus Humano 1/metabolismo , Interacciones Huésped-Patógeno , Humanos , Fusión de Membrana , Mutagénesis Sitio-Dirigida , Viroterapia Oncolítica , Receptores Virales/genética , Células Vero , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus
13.
Cancer Sci ; 107(6): 726-33, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27015293

RESUMEN

The programmed death-1/programmed death-1 ligands (PD-1/PD-L) pathway plays an important role in immunological tumor evasion. However, the clinical significance of the PD-L (L1 and L2) expression in esophageal cancer treated with chemotherapy has not been fully investigated. We examined the expression of PD-L of the primary tumors obtained from 180 esophageal cancer patients who underwent radical resection with or without neoadjuvant chemotherapy (NAC) using immunohistochemical staining. The relationship between the expression patterns and clinico-pathological characteristics was examined. In the present study, 53 patients (29.4%) and 88 patients (48.3%) were classified into positive for PD-L1 and PD-L2 expression, respectively. In all the patients examined, overall survival rates of the patients with tumors positive for PD-L1 or PD-L2 were significantly worse than those with tumors negative for PD-L1 or PD-L2 (P = 0.0010 and P = 0.0237, respectively). However, subgroup analysis showed that these tendencies are only found in the patients treated with NAC, and not in those without NAC. The patients with positive PD-L1 expression had a significantly higher rate of NAC history (P = 0.0139), but those with positive PD-L2 expression did not have a significantly high rate of NAC history (P = 0.6127). There is no significant relationship between PD-L1 expression and response to chemotherapy (P = 0.3118), but patients with positive PD-L2 expression had significantly inferior responses to chemotherapy (P = 0.0034). The PD-1/PD-L pathway might be an immunological mechanism associated with the long-term effectiveness of chemotherapy in esophageal cancer patients. Further investigation into the roles of PD-1 pathway in chemotherapy could lead to the development of better treatment options for this disease.


Asunto(s)
Antineoplásicos/uso terapéutico , Antígeno B7-H1/metabolismo , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/metabolismo , Humanos , Inmunohistoquímica , Ligandos , Persona de Mediana Edad , Terapia Neoadyuvante , Pronóstico , Tasa de Supervivencia
14.
Cancer Sci ; 107(9): 1206-14, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27384869

RESUMEN

Although it has been suspected that inflammation is associated with increased tumor metastasis, the exact type of immune response required to initiate cancer progression and metastasis remains unknown. In this study, by using an in vivo tumor progression model in which low tumorigenic cancer cells acquire malignant metastatic phenotype after exposure to inflammation, we found that IL-17A is a critical cue for escalating cancer cell malignancy. We further demonstrated that the length of exposure to an inflammatory microenvironment could be associated with acquiring greater tumorigenicity and that IL-17A was critical for amplifying such local inflammation, as observed in the production of IL-1ß and neutrophil infiltration following the cross-talk between cancer and host stromal cells. We further determined that γδT cells expressing Vδ1 semi-invariant TCR initiate cancer-promoting inflammation by producing IL-17A in an MyD88/IL-23-dependent manner. Finally, we identified CD30 as a key molecule in the inflammatory function of Vδ1T cells and the blockade of this pathway targeted this cancer immune-escalation process. Collectively, these results reveal the importance of IL-17A-producing CD30(+) Vδ1T cells in triggering inflammation and orchestrating a microenvironment leading to cancer progression.


Asunto(s)
Inflamación/inmunología , Inflamación/metabolismo , Interleucina-17/biosíntesis , Antígeno Ki-1/metabolismo , Neoplasias/etiología , Neoplasias/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inmunidad , Inflamación/complicaciones , Ratones , Ratones Noqueados , Modelos Biológicos , Neoplasias/patología , Microambiente Tumoral/inmunología
15.
Proc Natl Acad Sci U S A ; 109(6): 2066-71, 2012 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-22308357

RESUMEN

Smoldering inflammation often increases the risk of progression for malignant tumors and simultaneously matures myeloid dendritic cells (mDCs) for cell-mediated immunity. PolyI:C, a dsRNA analog, is reported to induce inflammation and potent antitumor immune responses via the Toll-like receptor 3/Toll-IL-1 receptor domain-containing adaptor molecule 1 (TICAM-1) and melanoma differentiation-associated protein 5/IFN-ß promoter stimulator 1 (IPS-1) pathways in mDCs to drive activation of natural killer cells and cytotoxic T lymphocytes. Here, we found that i.p. or s.c. injection of polyI:C to Lewis lung carcinoma tumor-implant mice resulted in tumor regression by converting tumor-supporting macrophages (Mfs) to tumor suppressors. F4/80(+)/Gr1(-) Mfs infiltrating the tumor respond to polyI:C to rapidly produce inflammatory cytokines and thereafter accelerate M1 polarization. TNF-α was increased within 1 h in both tumor and serum upon polyI:C injection into tumor-bearing mice, followed by tumor hemorrhagic necrosis and growth suppression. These tumor responses were abolished in TNF-α(-/-) mice. Furthermore, F4/80(+) Mfs in tumors extracted from polyI:C-injected mice sustained Lewis lung carcinoma cytotoxic activity, and this activity was partly abrogated by anti-TNF-α Ab. Genes for supporting M1 polarization were subsequently up-regulated in the tumor-infiltrating Mfs. These responses were completely abrogated in TICAM-1(-/-) mice, and unaffected in myeloid differentiation factor 88(-/-) and IPS-1(-/-) mice. Thus, the TICAM-1 pathway is not only important to mature mDCs for cross-priming and natural killer cell activation in the induction of tumor immunity, but also critically engaged in tumor suppression by converting tumor-supporting Mfs to those with tumoricidal properties.


Asunto(s)
Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Células Mieloides/inmunología , Células Mieloides/patología , Transducción de Señal/inmunología , Receptor Toll-Like 3/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Antineoplásicos/farmacología , Polaridad Celular/efectos de los fármacos , Polaridad Celular/inmunología , Citotoxicidad Inmunológica/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Ratones , Poli I-C/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
16.
Proc Natl Acad Sci U S A ; 108(30): 12425-30, 2011 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-21746895

RESUMEN

Recent evidence has unveiled the critical role of tumor cells with stem cell activities in tumorigenicity and drug resistance, but how tumor microenvironments regulate cancer stem/initiating cells (CSCs) remains unknown. We clarified the role of tumor-associated macrophages (TAMs) and their downstream factor milk-fat globule-epidermal growth factor-VIII (MFG-E8) in the regulation of CSC activities. Bone marrow chimeric systems and adoptive cell transfers elucidated the importance of MFG-E8 from TAMs in conferring to CSCs with the ability to promote tumorigenicity and anticancer drug resistance. MFG-E8 mainly activates signal transducer and activator of transcription-3 (Stat3) and Sonic Hedgehog pathways in CSCs and further amplifies their anticancer drug resistance in cooperation with IL-6. Thus, the pharmacological targeting of key factors derived from tumor-associated inflammation provides a unique strategy to eradicate therapy-resistant tumors by manipulating CSC activities.


Asunto(s)
Macrófagos/inmunología , Neoplasias/inmunología , Neoplasias/patología , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/patología , Traslado Adoptivo , Animales , Antígenos de Superficie/genética , Antígenos de Superficie/inmunología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Proteínas Hedgehog/inmunología , Humanos , Interleucina-6/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Proteínas de la Leche/genética , Proteínas de la Leche/inmunología , Modelos Inmunológicos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Células Madre Neoplásicas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Transducción de Señal/inmunología , Microambiente Tumoral/inmunología
17.
Cancer Med ; 13(3): e7042, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38400666

RESUMEN

BACKGROUND: Liver injury associated with oxaliplatin (L-OHP)-based chemotherapy can significantly impact the treatment outcomes of patients with colorectal cancer liver metastases, especially when combined with surgery. To date, no definitive biomarker that can predict the risk of liver injury has been identified. This study aimed to investigate whether organoids can be used as tools to predict the risk of liver injury. METHODS: We examined the relationship between the clinical signs of L-OHP-induced liver injury and the responses of patient-derived liver organoids in vitro. Organoids were established from noncancerous liver tissues obtained from 10 patients who underwent L-OHP-based chemotherapy and hepatectomy for colorectal cancer. RESULTS: Organoids cultured in a galactose differentiation medium, which can activate the mitochondria of organoids, showed sensitivity to L-OHP cytotoxicity, which was significantly related to clinical liver toxicity induced by L-OHP treatment. Organoids from patients who presented with a high-grade liver injury to the L-OHP regimen showed an obvious increase in mitochondrial superoxide levels and a significant decrease in mitochondrial membrane potential with L-OHP exposure. L-OHP-induced mitochondrial oxidative stress was not observed in the organoids from patients with low-grade liver injury. CONCLUSIONS: These results suggested that L-OHP-induced liver injury may be caused by mitochondrial oxidative damage. Furthermore, patient-derived liver organoids may be used to assess susceptibility to L-OHP-induced liver injury in individual patients.


Asunto(s)
Antineoplásicos , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas , Neoplasias Colorrectales , Humanos , Oxaliplatino/efectos adversos , Neoplasias Colorrectales/patología , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/tratamiento farmacológico , Organoides/patología , Antineoplásicos/efectos adversos
18.
Blood ; 117(9): 2640-8, 2011 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-21220748

RESUMEN

Dendritic cells (DCs) are known to regulate immune responses by inducing both central and peripheral tolerance. DCs play a vital role in negative selection of developing thymocytes by deleting T cells with high-affinity for self-peptide-major histocompatibility complexes. In the periphery, DCs mediate peripheral tolerance by promoting regulatory T-cell development, induction of T-cell unresponsiveness, and deletion of activated T cells. We studied whether allogeneic DCs, obtained from bone marrow cultured with either Flt3L (FLDCs) or granulocyte-macrophage colony-stimulating factor (GMDCs), could induce allospecific central and peripheral tolerance after IV injection; B cells were used as a control. The results showed that only FLDCs reached the thymus after injection and that these cells induced both central and peripheral tolerance to donor major histocompatibility complexes. For central tolerance, injection of FLDCs induced antigen-specific clonal deletion of both CD8 and CD4 single-positive thymocytes. For peripheral tolerance, injection of FLDCs induced donor-specific T-cell unresponsiveness and prolonged survival of donor-derived skin grafts. Tolerance induction by adoptive transfer of FLDCs could be a useful approach for promoting graft acceptance after organ transplantation.


Asunto(s)
Células Dendríticas/citología , Células Dendríticas/inmunología , Supervivencia de Injerto/inmunología , Tolerancia Inmunológica/inmunología , Trasplante de Piel/inmunología , Traslado Adoptivo , Animales , Movimiento Celular , Proliferación Celular , Células Clonales , Epítopos/inmunología , Inyecciones , Proteínas de la Membrana/metabolismo , Ratones , Células de Población Lateral/citología , Células de Población Lateral/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Timo/citología , Factores de Tiempo , Trasplante Homólogo
19.
J Appl Lab Med ; 8(6): 1065-1073, 2023 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-37748758

RESUMEN

BACKGROUND: Next-generation sequencing (NGS)-based genomic profiling is becoming widespread in determining treatment policies for patients with tumors. Commercially available gene panels for pan-tumor targets comprise hundreds of tumor-related genes but frequently lack genes of interest in specific tumor types. In this study, we demonstrate a method for extending target regions of genomic profiling by combining a custom probe pool with a commercial targeted panel. METHODS: We used TruSight Oncology 500 (TSO500) as a commercial targeted panel and a custom probe pool designed for all exons of the SMARCA2 gene. Sequencing libraries of custom targets were constructed using a portion of the TSO500 library solution before the hybridization-capture process. After hybridization capture, both libraries were combined and sequenced using a next-generation sequencer. RESULTS: Sequencing results showed that >96.8% and 100% of the target exons were covered at a depth of over 100× using the TSO500 and custom panels, respectively. The custom panels had slightly better median exon coverage than the TSO500. The combined libraries of the custom and TSO500 panels showed a mapped read ratio close to the mixing ratio. Analysis of mutation-free regions showed similar accuracies between the TSO500 and custom panels regarding variant calling. CONCLUSIONS: Our devised method easily and affordably extends the targets beyond a ready-made panel. This method provides a valuable solution until the widespread adoption of whole-exome sequencing, which is costly for large target sizes.


Asunto(s)
Neoplasias , Humanos , Mutación , Biblioteca de Genes , Neoplasias/genética , Genómica
20.
Mol Cancer Res ; 21(1): 76-85, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36162957

RESUMEN

IMPLICATIONS: Considering the importance of GSTA4 in controlling IFNγ responsiveness and the metastatic potential of other melanoma cells, our results highlight a novel mechanism whereby cancer cells escape from host immunity and gain metastatic ability by acquiring resistance to oxidative stress responses through the upregulation of GSTA4.


Asunto(s)
Interferón gamma , Melanoma , Humanos , Interferón gamma/genética , Melanoma/patología , Regulación hacia Arriba , Metástasis de la Neoplasia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA