Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
1.
J Pathol Clin Res ; 8(5): 458-469, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35762092

RESUMEN

Programmed cell death-1 (PD-1) and its ligand (PD-L1) are significant mediators of immune suppression in the tumor microenvironment. We focused on the immunological impact of PD-1/PD-L1 signaling during tumor progression in colorectal carcinoma (CRC) and its association with resistance to neoadjuvant chemoradiotherapy (NCRT) in locally advanced rectal carcinoma (LAd-RC). Histopathological and immunohistochemical analyses of 100 CRC cases (including 34 RC) without NCRT and 109 NCRT-treated LAd-RC cases were performed. Membranous tumoral PD-L1 expression was identified in 9 of 100 (9%) CRC cases, including 1 of 34 (2.9%) RC cases, but PD-L1 immunopositivity was not associated with any clinicopathological factors, with the exception of deficient mismatch repair (dMMR) status. In contrast, stromal PD-L1+ immune cells, which frequently exhibited coexpression of PD-1 and CD8 markers, were significantly correlated with tumor vessel invasion, nuclear ß-catenin+ tumor budding cancer stem cell (CSC)-like features, and unfavorable prognosis. In the LAd-RC cases, stromal CD8+ (but not PD-L1+) immune cell infiltration in pretreatment-biopsied samples was significantly and positively associated with therapeutic efficacy. After NCRT, tumoral PD-L1 expression was observed in only 2 of 83 (2.4%) tumors, independent of dMMR status, whereas high stromal PD-L1+ and tumoral nuclear ß-catenin positivity were significantly linked to a poor response to NCRT and high tumor budding features. In addition, high stromal PD-L1 immunoreactivity was significantly associated with poorer overall survival. In conclusion, a combination of stromal PD-L1+ immune cells and nuclear ß-catenin+ tumor budding may contribute to tumor progression in CRC and resistance to NCRT in LAd-RC, through formation of niche-like lesions that exhibit immune resistance and CSC properties.


Asunto(s)
Antígeno B7-H1 , Resistencia a Antineoplásicos , Receptor de Muerte Celular Programada 1 , Tolerancia a Radiación , Neoplasias del Recto , beta Catenina , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Núcleo Celular/genética , Núcleo Celular/inmunología , Quimioradioterapia Adyuvante , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/inmunología , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Terapia Neoadyuvante , Pronóstico , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Tolerancia a Radiación/genética , Tolerancia a Radiación/inmunología , Neoplasias del Recto/genética , Neoplasias del Recto/inmunología , Neoplasias del Recto/patología , Neoplasias del Recto/terapia , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , beta Catenina/genética , beta Catenina/inmunología
2.
J Extracell Vesicles ; 10(14): e12169, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34894384

RESUMEN

Despite advancements in treatments, oral squamous cell carcinoma (OSCC) has not significantly improved in prognosis or survival rate primarily due to the presence of treatment-resistant OSCC. The intercellular communication between tumour cells is a molecular mechanism involved in acquiring OSCC treatment resistance. Extracellular vesicles (EVs) and encapsulated miRNAs are important mediators of intercellular communication. Here, we focused on EVs released from clinically relevant radioresistant (CRR) OSCC cells. Additionally, we evaluated the correlation between miRNA expression in the serum samples of patients who showed resistance to radiotherapy and in EVs released from CRR OSCC cells. We found that EVs released from CRR OSCC cells conferred radioresistance to radiosensitive OSCC cells via miR-503-3p contained in EVs. This miR-503-3p inhibited BAK and impaired the caspase cascade to suppress radiation-induced apoptosis. Furthermore, OSCC cells with BAK knockdown had increased radioresistance. Additionally, the expression of circulating miR-503-3p in patients with OSCC was correlated with a poor treatment response and prognosis of radiotherapy. Our results provide new insights into the relationship between EVs and the radioresistance of OSCC and suggest that the miR-503-3p-BAK axis may be a therapeutic target and that circulating miR-503-3p is a useful prognostic biomarker in the radiotherapy of OSCC.


Asunto(s)
Carcinoma de Células Escamosas/radioterapia , Vesículas Extracelulares/metabolismo , MicroARNs/metabolismo , Neoplasias de la Boca/radioterapia , Tolerancia a Radiación/inmunología , Línea Celular Tumoral , Humanos , Transfección
3.
Sci Immunol ; 6(60)2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34723044

RESUMEN

Radiotherapy (RT) is an important anti-cancer treatment modality that activates innate and adaptive immune responses. When all-trans retinoic acid (RA) was administered with radiation, we observed superior antitumor responses compared to ionizing radiation (IR) alone or RA alone. The superior antitumor effects of combination treatment were accompanied by a dramatic increase of TNF-α- and inducible nitric oxide synthase (iNOS)-producing inflammatory macrophages in local and distal non-irradiated (distal) tumors. Inflammatory macrophages are essential for the therapeutic efficacy of combination treatment by inducing effector T cell infiltration and enhancing the effector T cell to regulatory T cell ratio in local and distal tumors. T cells and T cell-derived IFN-γ are crucial for increasing inflammatory macrophage levels in IR and RA treated tumors. Notably, whereas CD8+ T cells are required for the antitumor response to IR, CD4+ T cells are required for the effectiveness of the IR and RA combination. Combination treatment with RA enhanced the abscopal response when radiation and PD-L1 blockade were used together. The synergistic positive feedback loop of inflammatory macrophages and adaptive immunity is required for the antitumor efficacy of IR plus RA combination treatment. Our findings provide a translational and relatively nontoxic strategy for enhancing the local and systemic antitumor effects of IR.


Asunto(s)
Quimioradioterapia/métodos , Macrófagos/efectos de los fármacos , Neoplasias/terapia , Tretinoina/farmacología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Interferón gamma/genética , Interferón gamma/metabolismo , Macrófagos/inmunología , Ratones , Ratones Noqueados , Neoplasias/inmunología , Neoplasias/patología , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/inmunología , Receptores CCR2/genética , Receptores CCR2/metabolismo , Tretinoina/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación
4.
Neoplasia ; 23(11): 1110-1122, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34619428

RESUMEN

Radiotherapy efficacy is the result of radiation-mediated cytotoxicity coupled with stimulation of antitumor immune responses. We develop an in silico 3-dimensional agent-based model of diverse tumor-immune ecosystems (TIES) represented as anti- or pro-tumor immune phenotypes. We validate the model in 10,469 patients across 31 tumor types by demonstrating that clinically detected tumors have pro-tumor TIES. We then quantify the likelihood radiation induces antitumor TIES shifts toward immune-mediated tumor elimination by developing the individual Radiation Immune Score (iRIS). We show iRIS distribution across 31 tumor types is consistent with the clinical effectiveness of radiotherapy, and in combination with a molecular radiosensitivity index (RSI) combines to predict pan-cancer radiocurability. We show that iRIS correlates with local control and survival in a separate cohort of 59 lung cancer patients treated with radiation. In combination, iRIS and RSI predict radiation-induced TIES shifts in individual patients and identify candidates for radiation de-escalation and treatment escalation. This is the first clinically and biologically validated computational model to simulate and predict pan-cancer response and outcomes via the perturbation of the TIES by radiotherapy.


Asunto(s)
Biomarcadores/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/patología , Linfocitos Infiltrantes de Tumor/inmunología , Tolerancia a Radiación/genética , Microambiente Tumoral , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/radioterapia , Pronóstico , Tolerancia a Radiación/inmunología , Radioterapia , Tasa de Supervivencia
5.
Int J Mol Sci ; 22(19)2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-34638817

RESUMEN

Local radiotherapy (RT) is important to manage metastatic triple-negative breast cancer (TNBC). Although RT primarily reduces cancer cells locally, this control can be enhanced by triggering the immune system via immunotherapy. RT and immunotherapy may lead to an improved systemic effect, known as the abscopal effect. Here, we analyzed the antitumor effect of combination therapy using RT with an anti-programmed cell death-1 (PD-1) antibody in primary tumors, using poorly immunogenic metastatic mouse mammary carcinoma 4T1 model. Mice were injected subcutaneously into both flanks with 4T1 cells, and treatment was initiated 12 days later. Mice were randomly assigned to three treatment groups: (1) control (no treatment with RT or immune checkpoint inhibitor (ICI)), (2) RT alone, and (3) RT+ICI. The same RT dose was prescribed in both RT-alone and RT+ICI groups as 10Gy/fx in two fractions and delivered to only one of the two tumor burdens injected at both sides of flanks. In the RT+ICI group, 200 µg fixed dose of PD-1 antibody was intraperitoneally administered concurrently with RT. The RT and ICI combination markedly reduced tumor cell growth not only in the irradiated site but also in non-irradiated sites, a typical characteristic of the abscopal effect. This was observed only in radiation-sensitive cancer cells. Lung metastasis development was lower in RT-irradiated groups (RT-only and RT+ICI groups) than in the non-irradiated group, regardless of the radiation sensitivity of tumor cells. However, there was no additive effect of ICI on RT to control lung metastasis, as was already known regarding the abscopal effect. The combination of local RT with anti-PD-1 blockade could be a promising treatment strategy against metastatic TNBC. Further research is required to integrate our results into a clinical setting.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Pulmonares/prevención & control , Neoplasias Mamarias Experimentales/terapia , Tolerancia a Radiación/efectos de los fármacos , Animales , Línea Celular Tumoral , Femenino , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Tolerancia a Radiación/inmunología , Tolerancia a Radiación/efectos de la radiación
6.
Clin Immunol ; 229: 108776, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34118401

RESUMEN

Early-onset ataxia with ocular motor apraxia and hypoalbuminemia (EAOH) is a neurodegenerative disorder caused by mutation in the aprataxin (APTX)-coding gene APTX, which is involved in DNA single-strand break repair (SSBR). The neurological abnormalities associated with EAOH are similar to those observed in patients with ataxia-telangiectasia. However, the immunological abnormalities in patients with EAOH have not been described. In this study, we report that EAOH patients have immunological abnormalities, including lymphopenia; decreased levels of CD4+ T-cells, CD8+ T-cells, and B-cells; hypogammaglobulinemia; low T-cell recombination excision circles and kappa-deleting element recombination circles; and oligoclonality of T-cell receptor ß-chain variable repertoire. These immunological abnormalities vary among the EAOH patients. Additionally, mild radiosensitivity in the lymphocytes obtained from the patients with EAOH was demonstrated. These findings suggested that the immunological abnormalities and mild radiosensitivity evident in patients with EAOH could be probably caused by the DNA repair defects.


Asunto(s)
Apraxias/inmunología , Ataxia Cerebelosa/congénito , Hipoalbuminemia/inmunología , Adolescente , Adulto , Apraxias/genética , Apraxias/metabolismo , Estudios de Casos y Controles , Ataxia Cerebelosa/genética , Ataxia Cerebelosa/inmunología , Ataxia Cerebelosa/metabolismo , Niño , Roturas del ADN de Cadena Simple , Reparación del ADN/genética , Reparación del ADN/efectos de la radiación , Proteínas de Unión al ADN/genética , Femenino , Genes Codificadores de los Receptores de Linfocitos T , Variación Genética , Humanos , Hipoalbuminemia/genética , Hipoalbuminemia/metabolismo , Masculino , Persona de Mediana Edad , Mutación , Proteínas Nucleares/genética , Tolerancia a Radiación/genética , Tolerancia a Radiación/inmunología , Linfocitos T/inmunología , Adulto Joven
7.
Nat Commun ; 12(1): 3229, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-34050181

RESUMEN

Radiotherapy (RT)-induced tumoricidal immunity is severely limited when tumors are well-established. Here, we report that depleting SIRPα on intratumoral macrophages augments efficacy of RT to eliminate otherwise large, treatment-resistant colorectal (MC38) and pancreatic (Pan02 and KPC) tumors, inducing complete abscopal remission and long-lasting humoral and cellular immunity that prevent recurrence. SIRPα-deficient macrophages activated by irradiated tumor-released DAMPs exhibit robust efficacy and orchestrate an anti-tumor response that controls late-stage tumors. Upon RT-mediated activation, intratumoral SIRPα-deficient macrophages acquire potent proinflammatory features and conduct immunogenic antigen presentation that confer a tumoricidal microenvironment highly infiltrated by tumor-specific cytotoxic T cells, NK cells and inflammatory neutrophils, but with limited immunosuppressive regulatory T cells, myeloid derived suppressor cells and post-radiation wound-healing. The results demonstrate that SIRPα is a master regulator underlying tumor resistance to RT and provide proof-of-principle for SIRPα-deficient macrophage-based therapies to treat a broad spectrum of cancers, including those at advanced stages with low immunogenicity and metastases.


Asunto(s)
Neoplasias/terapia , Tolerancia a Radiación/inmunología , Receptores Inmunológicos/metabolismo , Linfocitos T Citotóxicos/inmunología , Macrófagos Asociados a Tumores/inmunología , Alarminas/inmunología , Alarminas/metabolismo , Alarminas/efectos de la radiación , Animales , Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoterapia/métodos , Masculino , Ratones , Ratones Noqueados , Neoplasias/inmunología , Neoplasias/patología , Prueba de Estudio Conceptual , Receptores Inmunológicos/genética , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/trasplante
8.
Front Immunol ; 12: 646384, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054811

RESUMEN

Inadequate sustained immune activation and tumor recurrence are major limitations of radiotherapy (RT), sustained and targeted activation of the tumor microenvironment can overcome this obstacle. Here, by two models of a primary rat breast cancer and cell co-culture, we demonstrated that valproic acid (VPA) and its derivative (HPTA) are effective immune activators for RT to inhibit tumor growth by inducing myeloid-derived macrophages and polarizing them toward the M1 phenotype, thus elevate the expression of cytokines such as IL-12, IL-6, IFN-γ and TNF-α during the early stage of the combination treatment. Meanwhile, activated CD8+ T cells increased, angiogenesis of tumors is inhibited, and the vasculature becomes sparse. Furthermore, it was suggested that VPA/HPTA can enhance the effects of RT via macrophage-mediated and macrophage-CD8+ T cell-mediated anti-tumor immunity. The combination of VPA/HPTA and RT treatment slowed the growth of tumors and prolong the anti-tumor effect by continuously maintaining the activated immune response. These are promising findings for the development of new effective, low-cost concurrent cancer therapy.


Asunto(s)
Neoplasias de la Mama/radioterapia , Linfocitos T CD8-positivos/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Tolerancia a Radiación/efectos de los fármacos , Ácido Valproico/farmacología , Animales , Neoplasias de la Mama/inmunología , Linfocitos T CD8-positivos/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Humanos , Inmunidad/efectos de los fármacos , Inmunidad/inmunología , Inmunidad/efectos de la radiación , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Tolerancia a Radiación/inmunología , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Transducción de Señal/efectos de la radiación , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación , Ácido Valproico/química
9.
Nat Commun ; 12(1): 2776, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33986291

RESUMEN

Radiation therapy (RT) is used in the management of several cancers; however, tumor radioresistance remains a challenge. Polymorphonuclear neutrophils (PMNs) are recruited to the tumor immune microenvironment (TIME) post-RT and can facilitate tumor progression by forming neutrophil extracellular traps (NETs). Here, we demonstrate a role for NETs as players in tumor radioresistance. Using a syngeneic bladder cancer model, increased NET deposition is observed in the TIME of mice treated with RT and inhibition of NETs improves overall radiation response. In vitro, the protein HMGB1 promotes NET formation through a TLR4-dependent manner and in vivo, inhibition of both HMGB1 and NETs significantly delays tumor growth. Finally, NETs are observed in bladder tumors of patients who did not respond to RT and had persistent disease post-RT, wherein a high tumoral PMN-to-CD8 ratio is associated with worse overall survival. Together, these findings identify NETs as a potential therapeutic target to increase radiation efficacy.


Asunto(s)
Trampas Extracelulares/metabolismo , Neutrófilos/inmunología , Tolerancia a Radiación/inmunología , Neoplasias de la Vejiga Urinaria/radioterapia , Anciano , Anciano de 80 o más Años , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Femenino , Proteína HMGB1/antagonistas & inhibidores , Proteína HMGB1/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Microambiente Tumoral/inmunología , Neoplasias de la Vejiga Urinaria/patología
10.
Biomed Res Int ; 2021: 6653747, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33681363

RESUMEN

OBJECTIVE: This study is set out to determine the relationship between IL-32 and radiosensitivity of esophageal squamous cell carcinoma (ESCC). METHODS: Western blot was adopted for measuring IL-32 expression in Eca-109 and TE-10 cells. Eca-109 and TE-10 cells with interference or overexpression of IL-32 were treated with the presence or absence of X-ray irradiation. Then, the use of CCK8 assay was to detect proliferation ability, and effects of IL-32 expression on radiosensitivity of ESCC were tested by colony formation assay. The cell apoptosis was detected using flow cytometry. STAT3 and p-STAT expression, and apoptotic protein Bax were detected by western blot. RESULTS: Colony formation assay and CCK8 assay showed that compared with the NC group without treatment, the growth of the ESCC cells, that is Eca-109 and TE-10, was significantly inhibited in the OE+IR group with highly expressed IL-32 and irradiation. In flow cytometry analysis, in Eca-109 and TE-10 cells, highly expressed IL-32 combined with irradiation significantly increased apoptosis compared with the control group. Highly expressed IL-32 has a synergistic effect with irradiation, inhibiting STAT3 and p-STAT3 expression and increasing apoptotic protein Bax expression. CONCLUSION: IL-32 can improve the radiosensitivity of ESCC cells by inhibiting the STAT3 pathway. Therefore, IL-32 can be used as a new therapeutic target to provide a new attempt for radiotherapy of ESCC.


Asunto(s)
Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Interleucinas/inmunología , Proteínas de Neoplasias/inmunología , Tolerancia a Radiación/inmunología , Factor de Transcripción STAT3/inmunología , Transducción de Señal/inmunología , Línea Celular Tumoral , Neoplasias Esofágicas/inmunología , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/radioterapia , Carcinoma de Células Escamosas de Esófago/inmunología , Carcinoma de Células Escamosas de Esófago/patología , Carcinoma de Células Escamosas de Esófago/radioterapia , Humanos
11.
J Immunol Res ; 2021: 6657012, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33506057

RESUMEN

BACKGROUND: Preoperative chemoradiotherapy (pCRT) is a common and essential therapeutic strategy for patients with locally advanced rectal cancer (LARC), but poor tumor response and therapeutic resistance to chemoradiotherapy have appeared usually among persons and affected those patients' survival prognosis. The resistance to chemoradiotherapy in rectal cancer is difficult to predict. This study was aimed at evaluating the role of V-set and transmembrane domain containing 2 like protein (VSTM2L) in resistance to chemoradiotherapy in rectal cancer. METHODS: Analysis of the GEO profiling datasets of rectal cancer patients receiving pCRT disclosed that VSTM2L as a candidate gene was significantly upregulated in nonresponders of rectal cancer with pCRT. The mRNA and protein expression of VSTM2L was detected by quantitate real-time PCR, western blotting, and immunohistochemistry in six rectal cancer biopsy tissues before pCRT. Furthermore, the rectal cancer patient-derived organoids were cultured to evaluate the association of VSTM2L expression and tumor response to CRT. Overexpression of VSTM2L in cancer cells treated with CRT was analyzed for the function of cell proliferation and viability, clone formation, DNA damage repair, and apoptosis ability. The GSEA and RNA-sequence analysis were used to find the downstream mechanism of VSTM2L overexpression in cells treated with CRT. RESULTS: The mRNA levels of VSTM2L were significantly downregulated in normal rectal tissues compared to tumor tissues and were upregulated in nonresponders of rectal cancer patients receiving pCRT and positively correlated with poor survival prognosis from GEO datasets. High expression of VSTM2L was significantly associated with tumor regression after pCRT (P = 0.030). Moreover, high expression of VSTM2L reduced γ-H2AX expression in rectal cancer patient-derived organoids treated with CRT. The overexpression of VSTM2L in colorectal cancer cells induced resistance to CRT via promoting cell proliferation and inhibiting apoptosis. The molecular mechanism revealed that the overexpression of VSTM2L induced resistance to CRT through downstream IL-4 signaling affecting the progress of cell proliferation and apoptosis. CONCLUSION: The high expression of VSTM2L induced resistance to CRT, and adverse survival outcomes served as a prognostic factor in patients with rectal cancer receiving pCRT, suggesting that VSTM2L high expression may be a potential resistant predictable biomarker for LARC patients receiving pCRT.


Asunto(s)
Quimioradioterapia/estadística & datos numéricos , Terapia Neoadyuvante/estadística & datos numéricos , Recurrencia Local de Neoplasia/epidemiología , Proteínas del Tejido Nervioso/metabolismo , Neoplasias del Recto/terapia , Adulto , Anciano , Línea Celular Tumoral , Conjuntos de Datos como Asunto , Supervivencia sin Enfermedad , Regulación hacia Abajo , Resistencia a Antineoplásicos/inmunología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Interleucina-4/metabolismo , Masculino , Persona de Mediana Edad , Terapia Neoadyuvante/métodos , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/prevención & control , Proctectomía , Pronóstico , Tolerancia a Radiación/inmunología , Neoplasias del Recto/genética , Neoplasias del Recto/inmunología , Neoplasias del Recto/mortalidad , Transducción de Señal/inmunología
12.
Int J Radiat Oncol Biol Phys ; 110(2): 510-520, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33383126

RESUMEN

PURPOSE: The establishment of a preclinical model of the abscopal effect on hepatocellular carcinoma (HCC) and evaluation of whether the hypofractionated radiation therapy (RT) multitumor Hepa1-6 mouse HCC model could be used to suppress nonradiated tumor mass was performed in this study. METHODS AND MATERIALS: Hepa1-6 mouse liver cancer cell lines were used to form tumors. Immunogenicity was analyzed using ELISpot and immune cell labeled antibody. Interferon (IFN) ß expression was confirmed through polymerase chain reaction. RESULTS: After investigation, the intratumoral transcription of type Ⅰ IFN increased by 2-fold. The antitumor immune response to Hepa 1-6 cells induced by radiation was increased. Moreover, the influx of activated CD8+ T cells was increased in nonirradiated tumors. The number of dendritic cells and activation status were evaluated by flow cytometry on the second day after irradiation. Flow cytometry revealed a significantly increased dendritic cell population expressing the CD11c molecule in tumor-draining lymph nodes. Furthermore, because irradiation leads to adaptation of immune resistance of tumor cells against RT, we sought to elucidate a potent tool to overcome the resistance and confirm the ability of PD-L1 antibody to survive late RT resistance. CONCLUSIONS: The immunologic mechanism of the abscopal effect was revealed and the application of PD-L1 inhibitor successfully performed as a breakthrough in late RT resistance in the Hepa1-6 tumor model.


Asunto(s)
Carcinoma Hepatocelular/terapia , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Hepáticas/terapia , Tolerancia a Radiación/inmunología , Radiocirugia/métodos , Animales , Antineoplásicos , Antígeno B7-H1/administración & dosificación , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Antígenos CD11/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Terapia Combinada/métodos , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/efectos de la radiación , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Interferón Tipo I/metabolismo , Interferón beta/metabolismo , Interferón gamma/inmunología , Interferón gamma/metabolismo , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Ganglios Linfáticos/metabolismo , Linfocitos Infiltrantes de Tumor/citología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Hipofraccionamiento de la Dosis de Radiación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Carga Tumoral/efectos de la radiación
13.
Br J Cancer ; 124(4): 817-830, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33214684

RESUMEN

BACKGROUND: Interferon (IFN) signalling pathways, a key element of the innate immune response, contribute to resistance to conventional chemotherapy, radiotherapy, and immunotherapy, and are often deregulated in cancer. The deubiquitylating enzyme USP18 is a major negative regulator of the IFN signalling cascade and is the predominant human protease that cleaves ISG15, a ubiquitin-like protein tightly regulated in the context of innate immunity, from its modified substrate proteins in vivo. METHODS: In this study, using advanced proteomic techniques, we have significantly expanded the USP18-dependent ISGylome and proteome in a chronic myeloid leukaemia (CML)-derived cell line. USP18-dependent effects were explored further in CML and colorectal carcinoma cellular models. RESULTS: Novel ISGylation targets were characterised that modulate the sensing of innate ligands, antigen presentation and secretion of cytokines. Consequently, CML USP18-deficient cells are more antigenic, driving increased activation of cytotoxic T lymphocytes (CTLs) and are more susceptible to irradiation. CONCLUSIONS: Our results provide strong evidence for USP18 in regulating antigenicity and radiosensitivity, highlighting its potential as a cancer target.


Asunto(s)
Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/inmunología , Citocinas/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/inmunología , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinas/metabolismo , Variación Antigénica , Línea Celular Tumoral , Neoplasias Colorrectales/radioterapia , Técnicas de Inactivación de Genes , Células HCT116 , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/radioterapia , Tolerancia a Radiación/genética , Tolerancia a Radiación/inmunología , Ubiquitina Tiolesterasa/deficiencia , Ubiquitina Tiolesterasa/genética
14.
Clin Cancer Res ; 27(3): 749-758, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33148672

RESUMEN

PURPOSE: Tumor-infiltrating immune cells play a key role in tumor progression. The purpose of this study was to analyze whether the immune infiltrate predicts benefit from postoperative radiotherapy in a large randomized breast cancer radiotherapy trial. EXPERIMENTAL DESIGN: In the SweBCG91RT trial, patients with stage I and II breast cancer were randomized to breast-conserving surgery (BCS) and postoperative radiotherapy or to BCS only and followed for a median time of 15.2 years. The primary tumor immune infiltrate was quantified through two independent methods: IHC and gene expression profiling. For IHC analyses, the absolute stromal area occupied by CD8+ T cells and FOXP3+ T cells, respectively, was used to define the immune infiltrate. For gene expression analyses, immune cells found to be prognostic in independent datasets were pooled into two groups consisting of antitumoral and protumoral immune cells, respectively. RESULTS: An antitumoral immune response in the primary tumor was associated with a reduced risk of breast cancer recurrence and predicted less benefit from adjuvant radiotherapy. The interaction between radiotherapy and immune phenotype was significant for any recurrence in both the IHC and gene expression analyses (P = 0.039 and P = 0.035) and was also significant for ipsilateral breast tumor recurrence in the gene expression analyses (P = 0.025). CONCLUSIONS: Patients with an antitumoral immune infiltrate in the primary tumor have a reduced risk of any recurrence and may derive less benefit from adjuvant radiotherapy. These results may impact decisions regarding postoperative radiotherapy in early breast cancer.


Asunto(s)
Neoplasias de la Mama/terapia , Mama/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Recurrencia Local de Neoplasia/epidemiología , Radioterapia Adyuvante , Adulto , Anciano , Mama/patología , Mama/efectos de la radiación , Mama/cirugía , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Linfocitos T CD8-positivos/inmunología , Femenino , Estudios de Seguimiento , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/inmunología , Humanos , Inmunohistoquímica , Linfocitos Infiltrantes de Tumor/metabolismo , Mastectomía Segmentaria , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/prevención & control , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Tolerancia a Radiación/genética , Tolerancia a Radiación/inmunología , Medición de Riesgo/métodos , Medición de Riesgo/estadística & datos numéricos , Análisis de Matrices Tisulares , Microambiente Tumoral/inmunología
15.
Oncol Rep ; 44(5): 2253-2264, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33000219

RESUMEN

Breast cancer is the most commonly diagnosed cancer and is the second leading cause of death in women. However, resistance to radio­ and chemotherapy remains one of the major difficulties in the treatment of breast cancer. Therefore, the aim of the present study was to identify novel regimens to overcome treatment resistance in patients with breast cancer. The results of the present study demonstrated that the attenuated Edmonston­B vaccine strain of the measles virus (MV­Edm) significantly re­sensitized breast cancer cells to doxorubicin and ionizing radiation. Mechanistically, MV­Edm reduced DNA double strand repair efficiency by decreasing the mRNA and protein expression levels of p53­binding protein 1 and disassembling the non­homologous end joining (NHEJ) complex. NHEJ deficiency, which was achieved using DNA ligase IV knockout via CRISPR/Cas9, resulted in failure to overcome resistance mediated by MV­Edm infection. As a result of the significant synergy between attenuated MV and radio­ or chemotherapy, MV­Edm provides a novel strategy for the treatment of radio­ and chemoresistant breast cancer.


Asunto(s)
Neoplasias de la Mama/terapia , Vacunas contra el Cáncer/administración & dosificación , Quimioradioterapia/métodos , Doxorrubicina/farmacología , Vacuna Antisarampión/administración & dosificación , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Vacunas contra el Cáncer/inmunología , Chlorocebus aethiops , Reparación del ADN por Unión de Extremidades/inmunología , ADN Ligasa (ATP)/genética , ADN Ligasa (ATP)/metabolismo , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos/inmunología , Femenino , Técnicas de Inactivación de Genes , Humanos , Células MCF-7 , Vacuna Antisarampión/inmunología , Tolerancia a Radiación/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Células Vero
16.
Oncol Rep ; 44(2): 698-710, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32627036

RESUMEN

The aim of the present study was to examine the potential role of human heparin­binding epidermal growth factor (HB­EGF) secreted by M2 macrophages in the development of radioresistance in head and neck squamous cell carcinoma (HNSCC). Immunohistochemistry was used to detect radiosensitivity in human papilloma virus (HPV)­positive and HPV­negative HNSCC tissues and immunohistochemical staining with specific antibodies for macrophage surface markers was used to assess the infiltration of M1 and M2 macrophages in HPV­positive and ­negative HNSCC tissues. The expression of HB­EGF in HPV­positive and ­negative HNSCC tissues was determined by multi­cytokine detection in order to determine the relationship between HB­EGF and radiosensitivity. M1 and M2 macrophages were co­cultured with the HNSCC cell line CAL27 and treated with HB­EGF and its neutralizing antibodies to assess radiation sensitivity. Finally, the major DNA double­strand break repair pathways required for the activation of HB­EGF and promotion of epidermal growth factor receptor (EGFR) were identified. The results revealed that radiosensitivity was higher in HPV­positive HNSCC compared with HPV­negative. There was a higher infiltration of M2 macrophages in HPV­negative HNSCC, which were revealed as the main source of HB­EGF secretion. Furthermore, it was determined that overexpression of HB­EGF induced radioresistance in HPV­negative HNSCC. HB­EGF promoted the activation of the non­homologous end­joining pathway by activating EGFR. To the best of our knowledge, this is the first study to demonstrate the association between HB­EGF and radiosensitivity in HNSCC. These results indicated that the secretion of HB­EGF by M2 macrophages could induce radioresistance of HPV­negative HNSCC.


Asunto(s)
Neoplasias de Cabeza y Cuello/radioterapia , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Macrófagos/inmunología , Infecciones por Papillomavirus/radioterapia , Tolerancia a Radiación/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/radioterapia , Línea Celular Tumoral , Técnicas de Cocultivo , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN por Unión de Extremidades , Receptores ErbB/metabolismo , Femenino , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/patología , Neoplasias de Cabeza y Cuello/virología , Humanos , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Papillomaviridae/aislamiento & purificación , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/virología
17.
Pathol Oncol Res ; 26(4): 2459-2467, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32564263

RESUMEN

Undifferentiated Nasopharyngeal Carcinoma (UNPC) is associated with Epstein-Barr Virus (EBV) and characterized by an abundant immune infiltrate potentially influencing the prognosis. Thus, we retrospectively assessed the significance of immunosuppression in the UNPC microenvironment as prognostic biomarker of treatment failure in a non-endemic area, and monitored the variation of systemic EBV-specific immunity before and after chemoradiotherapy (CRT). DNA and RNA were extracted from diagnostic biopsies obtained by tumor and adjacent mucosa from 63 consecutive EBV+ UNPC patients who underwent radical CRT. Among these patients 11 relapsed within 2 years. The expression of the EBV-derived UNPC-specific BARF1 gene and several immune-related genes was monitored through quantitative RT-PCR and methylation-specific PCR analyses. Peripheral T cell responses against EBV and BARF1 were measured in 14 patients (7 relapses) through IFN-γ ELISPOT assay. We found significantly higher expression levels of BARF1, CD8, IFN-γ, IDO, PD-L1, and PD-1 in UNPC samples compared to healthy tissues. CD8 expression was significantly reduced in both tumor and healthy tissues in UNPC patients who relapsed within two years. We observed a hypomethylated FOXP3 intron 1 exclusively in relapsed UNPC patients. Finally, we noticed a significant decrease in EBV- and BARF1-specific T-cells after CRT only in relapsing patients. Our data suggest that a high level of immunosuppression (low CD8, hypomethylated FoxP3) in UNPC microenvironment may predict treatment failure and may allow an early identification of patients who could benefit from the addition of immune modulating strategies to improve first line CRT.


Asunto(s)
Antígenos CD8/inmunología , Resistencia a Antineoplásicos/inmunología , Factores de Transcripción Forkhead/inmunología , Carcinoma Nasofaríngeo/inmunología , Neoplasias Nasofaríngeas/inmunología , Tolerancia a Radiación/inmunología , Adolescente , Adulto , Anciano , Quimioradioterapia/métodos , Metilación de ADN , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/inmunología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Carcinoma Nasofaríngeo/terapia , Neoplasias Nasofaríngeas/terapia , Valor Predictivo de las Pruebas , Estudios Retrospectivos , Microambiente Tumoral/inmunología , Proteínas Virales/inmunología , Adulto Joven
18.
Sci Rep ; 10(1): 2716, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-32066820

RESUMEN

A growing proportion of head and neck cancers (HNC) result from HPV infection. Between HNC aetiological groups (HPV positive and HPV negative) clinical evidence demonstrates significantly better treatment response among HPV positive cancers. Cancer stem cells (CSCs) are identified in HNC tumour populations as agents of treatment resistance and a target for tumour control. This study examines dynamic responses in populations of a CSC phenotype in HNC cell lines following X-irradiation at therapeutic levels, and comparing between HPV statuses. Variations in CSC density between HPV groups showed no correlation with better clinical outcomes seen in the HPV positive status. CSC populations in HPV positive cell lines ranged from 1.9 to 4.8%, and 2.6 to 9.9% for HPV negative. Following 4 Gy X- irradiation however, HPV negative cell lines demonstrated more frequent and significantly greater escalation in CSC proportions, being 3-fold that of the HPV positive group at 72 hours post irradiation. CSC proportions of tumour populations are not fixed but subject to change in response to radiation at therapeutic dose levels. These findings imply a potential effect of aetiology on radio-responsiveness in CSCs, illustrating that clonogen treatment response may be more informative of therapy outcomes than inherent population density alone.


Asunto(s)
Aldehído Deshidrogenasa/genética , Receptores de Hialuranos/genética , Células Madre Neoplásicas/efectos de la radiación , Papillomaviridae/patogenicidad , Tolerancia a Radiación/genética , Anciano , Aldehído Deshidrogenasa/inmunología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/radioterapia , Recuento de Células , Línea Celular Tumoral , Femenino , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/patología , Neoplasias de Cabeza y Cuello/radioterapia , Humanos , Receptores de Hialuranos/inmunología , Masculino , Persona de Mediana Edad , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/patología , Papillomaviridae/crecimiento & desarrollo , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/radioterapia , Tolerancia a Radiación/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/radioterapia , Rayos X
19.
Future Oncol ; 16(5): 103-115, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31933380

RESUMEN

Aim: To evaluate the predictive significance of systemic immune-inflammation index (SII) on overall survival (OS) and radiosensitivity in advanced non-small-cell lung cancer. Materials & methods: Kaplan-Meier analysis and Cox proportional hazard models were used to assess the prognostic value of SII. Results: The optimal cutoff for SII was 555.59, with an area under the curve of 0.782 (sensitivity: 76.6%, specificity: 71.9%, 95% CI: 0.730-0.833), respectively. Median OS (p < 0.001) in the low SII group (32.8 months) was better than the OS in the high SII group (8.5 months). SII-low group statistically exhibited a better radiosensitivity. Conclusion: SII was an independent prognostic factor for OS and predictive factor for radiosensitivity. Higher level of SII associated with poorer OS and poorer radiosensitivity.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Inflamación/radioterapia , Neutrófilos/metabolismo , Tolerancia a Radiación/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Pulmón de Células no Pequeñas/sangre , Carcinoma de Pulmón de Células no Pequeñas/complicaciones , Carcinoma de Pulmón de Células no Pequeñas/patología , Supervivencia sin Enfermedad , Femenino , Humanos , Inflamación/sangre , Inflamación/complicaciones , Inflamación/patología , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neutrófilos/patología , Pronóstico , Modelos de Riesgos Proporcionales
20.
Front Immunol ; 11: 581111, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33519804

RESUMEN

Postoperative ileus (POI) is triggered by an innate immune response in the muscularis externa (ME) and is accompanied by bacterial translocation. Bacteria can trigger an innate immune response via toll-like receptor (TLR) activation, but the latter's contribution to POI has been disproved for several TLRs, including TLR2 and TLR4. Herein we investigated the role of double-stranded RNA detection via TLR3 and TIR-domain-containing adapter-inducing interferon-ß (TRIF) signaling pathway in POI. POI was induced by small bowel intestinal manipulation in wt, TRIF-/-, TLR3-/-, type I interferon receptor-/- and interferon-ß reporter mice, all on C57BL/6 background, and POI severity was quantified by gene expression analysis, gastrointestinal transit and leukocyte extravasation into the ME. TRIF/TLR3 deficiency reduced postoperative ME inflammation and prevented POI. With bone marrow transplantation, RNA-sequencing, flow cytometry and immunohistochemistry we revealed a distinct TLR3-expressing radio-resistant MHCIIhiCX3CR1- IBA-1+ resident macrophage population within the deep myenteric plexus. TLR3 deficiency in these cells, but not in MHCIIhiCX3CR1+ macrophages, reduced cytokine expression in POI. While this might not be an exclusive macrophage-privileged pathway, the TLR3/TRIF axis contributes to proinflammatory cytokine production in MHCIIhiCX3CR1- IBA-1+ macrophages during POI. Deficiency in TLR3/TRIF protects mice from POI. These data suggest that TLR3 antagonism may prevent POI in humans.


Asunto(s)
Ileus/etiología , Macrófagos/inmunología , Complicaciones Posoperatorias/etiología , Receptor Toll-Like 3/inmunología , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Animales , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/inmunología , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Ileus/inmunología , Ileus/patología , Inmunidad Innata , Macrófagos/clasificación , Macrófagos/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Plexo Mientérico/inmunología , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/patología , Tolerancia a Radiación/inmunología , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 3/deficiencia , Receptor Toll-Like 3/genética , Quimera por Trasplante/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...