Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Br J Cancer ; 129(6): 935-946, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37537255

RESUMEN

BACKGROUND: Anti-oestrogenic therapy has been used for breast cancer patients with oestrogen susceptibility cancer cells. However, little has been known about its potential role for immune cell biology within TME, particularly in cancer patients without oestrogen sensitivity of tumour cells. Therefore, we aimed to study the effect of oestrogen on immunity within TME. METHODS: Using a clinical dataset, immune cells of humans and mice, female mice with and without ovaries, and several murine ERα-negative cancer cell lines, we evaluated the effect of oestrogen on immunity in TME. RESULTS: Clinical data analysis suggested oestrogen's suppressive efficacy against CTLs. Additionally, in vitro and in vivo experiments revealed intra-tumoural CTLs' direct repressive action by oestrogen in both mice and humans; blockade of oestrogen signals cancelled its immunosuppression resulting in tumour growth reduction in vivo. Most notably, immunotherapy (immune checkpoint inhibitor; ICI) combined with anti-oestrogenic therapy exhibited a dramatic anti-tumour effect. CONCLUSIONS: This study provides novel insights into how oestrogen contributes to tumour progression and a therapeutic rationale for blocking oestrogen signalling to boost the anti-tumour effect of ICI, regardless of tumour cells' ERα expression.


Asunto(s)
Neoplasias de la Mama , Receptor alfa de Estrógeno , Humanos , Femenino , Ratones , Animales , Receptor alfa de Estrógeno/metabolismo , Estrógenos/farmacología , Estrógenos/uso terapéutico , Neoplasias de la Mama/patología , Receptores de Estrógenos/metabolismo , Transducción de Señal
2.
Oncoimmunology ; 12(1): 2213132, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37235066

RESUMEN

Among cancer immunotherapy, which has received great attention in recent years, cancer vaccines can potentially prevent recurrent tumors by using the exquisite power and specificity of the immune system. Specifically, whole tumor cell vaccines (WTCVs) based on surgically resected tumors have been considered to elicit robust anti-tumor immune responses by exposing various tumor-associated antigens to host immunity. However, most tumors have little immunogenicity because of immunoediting by continuous interactions with host immunity; thus, preparing WTCVs based on patient-derived non-modified tumors cannot prevent tumor onset. Hence, the immunogenicity of tumor cells must be improved for effective WTCVs. In this study, we indicate the importance of the interferon regulatory factor 7 (Irf7) axis, including Irf7 and its downstream factors, within tumor cells in regulating immunogenicity. Indeed, WTCVs that augmented the Irf7 axis have exerted remarkable recurrence-preventive effects when vaccinated after tumor inactivation by radiation. Most notably, vaccination with murine colon cancer cells that enhanced the Irf7 axis prevented the development of challenged tumors in all mice and resulted in a 100% survival rate during the observation period. Furthermore, the mechanism leading to vaccine effectiveness was mediated by interferon-gamma-producing B cells. This study provides novel insights into how to enhance tumor immunogenicity and use WTCVs as recurrence prophylaxis.


Asunto(s)
Vacunas contra el Cáncer , Interferón gamma , Animales , Ratones , Recurrencia Local de Neoplasia/prevención & control , Factor 7 Regulador del Interferón/genética , Vacunas contra el Cáncer/farmacología , Antígenos de Neoplasias
3.
J Gynecol Oncol ; 34(3): e25, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36603850

RESUMEN

OBJECTIVE: Breast cancer susceptibility gene 1 (BRCA1)-associated ovarian cancer patients have been treated with A poly (ADP-ribose) polymerase (PARP) inhibitor, extending the progression-free survival; however, they finally acquire therapeutic resistance. Interleukin (IL)-34 has been reported as a poor prognostic factor in several cancers, including ovarian cancer, and it contributes to the therapeutic resistance of chemotherapies. IL-34 may affect the therapeutic effect of PARP inhibitor through the regulation of tumor microenvironment (TME). METHODS: In this study, The Cancer Genome Atlas (TCGA) data set was used to evaluate the prognosis of IL-34 and human ovarian serous carcinoma. We also used CRISPR-Cas9 genome editing technology in a mouse model to evaluate the efficacy of PARP inhibitor therapy in the presence or absence of IL-34. RESULTS: We found that IL34 was an independent poor prognostic factor in ovarian serous carcinoma, and its high expression significantly shortens overall survival. Furthermore, in BRCA1-associated ovarian cancer, PARP inhibitor therapy contributes to anti-tumor immunity via the XCR1+ DC-CD8+ T cell axis, however, it is canceled by the presence of IL-34. CONCLUSION: These results suggest that tumor-derived IL-34 benefits tumors by creating an immunosuppressive TME and conferring PARP inhibitor therapeutic resistance. Thus, we showed the pathological effect of IL-34 and the need for it as a therapeutic target in ovarian cancer.


Asunto(s)
Antineoplásicos , Carcinoma , Neoplasias Ováricas , Femenino , Animales , Ratones , Humanos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Interleucinas/genética , Interleucinas/uso terapéutico , Carcinoma/tratamiento farmacológico , Microambiente Tumoral
4.
Cancer Immunol Immunother ; 72(4): 851-864, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36104597

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype characterized by a lack of therapeutic targets. The paucity of effective treatment options motivated a number of studies to tackle this problem. Immunosuppressive cells infiltrated into the tumor microenvironment (TME) of TNBC are currently considered as candidates for new therapeutic targets. Myeloid-derived suppressor cells (MDSCs) have been reported to populate in the TME of TNBC, but their roles in the clinical and biological features of TNBC have not been clarified. This study identified that interleukin-34 (IL-34) released by TNBC cells is a crucial immunomodulator to regulate MDSCs accumulation in the TME. We provide evidence that IL-34 induces a differentiation of myeloid stem cells into monocytic MDSCs (M-MDSCs) that recruits regulatory T (Treg) cells, while suppressing a differentiation into polymorphonuclear MDSCs (PMN-MDSCs). As a result, the increase in M-MDSCs contributes to the creation of an immunosuppressive TME, and the decrease in PMN-MDSCs suppresses angiogenesis, leading to an acquisition of resistance to chemotherapy. Accordingly, blockade of M-MDSC differentiation with an estrogen receptor inhibitor or anti-IL-34 monoclonal antibody suppressed M-MDSCs accumulation causing retardation of tumor growth and restores chemosensitivity of the tumor by promoting PMN-MDSCs accumulation. This study demonstrates previously poorly understood mechanisms of MDSCs-mediated chemoresistance in the TME of TNBC, which is originated from the existence of IL-34, suggesting a new rationale for TNBC treatment.


Asunto(s)
Células Supresoras de Origen Mieloide , Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral , Linfocitos T Reguladores/patología , Interleucinas
6.
Rinsho Ketsueki ; 62(8): 1302-1307, 2021.
Artículo en Japonés | MEDLINE | ID: mdl-34497220

RESUMEN

The tumor microenvironment (TME) acquires immune resistance during the process of tumor formation. Recently, cancer immunotherapy has been attracting attention as a treatment modality, following the three major standard cancer treatments (surgical therapy, radiation therapy, and chemotherapy), for its potential to overcome such an immunosuppressive TME. Particularly, blocking antibodies against immune checkpoint molecules, such as PD-1 and CTLA-4 have caused a paradigm shift in cancer treatment. However, several patients do not respond to existing cancer immunotherapy; therefore, the establishment of a novel therapeutic target is essential. Macrophages are the most abundant cells in various tumors and are biased toward immunosuppressive forms. Therefore, research is ongoing globally to determine whether macrophages could be therapeutic targets. Interleukin-34 (IL-34) has been reported as a factor that biases macrophages to immunosuppressive forms. It is expressed in various types of cancer cells and plays important roles in multiple aspects of the TME. In this review, we comprehensively introduce the roles of IL-34 in the TME.


Asunto(s)
Neoplasias , Microambiente Tumoral , Humanos , Inmunoterapia , Interleucinas , Macrófagos , Neoplasias/terapia
7.
iScience ; 23(10): 101584, 2020 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-33205010

RESUMEN

Interleukin-34 (IL-34) is an alternative ligand to colony-stimulating factor-1 (CSF-1) for the CSF-1 receptor that acts as a key regulator of monocyte/macrophage lineage. In this study, we show that tumor-derived IL-34 mediates resistance to immune checkpoint blockade regardless of CSF-1 existence in various murine cancer models. Consistent with its immunosuppressive characteristics, the expression of IL-34 in tumors correlates with decreased frequencies of cellular (such as CD8+ and CD4+ T cells and M1-biased macrophages) and molecular (including various cytokines and chemokines) effectors at the tumor microenvironment. Then, a neutralizing antibody against IL-34 improved the therapeutic effects of the immune checkpoint blockade in combinatorial therapeutic models, including a patient-derived xenograft model. Collectively, we revealed that tumor-derived IL-34 inhibits the efficacy of immune checkpoint blockade and proposed the utility of IL-34 blockade as a new strategy for cancer therapy.

8.
Breast Cancer ; 27(6): 1198-1204, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32578004

RESUMEN

Triple-negative breast cancer (TNBC) is a subtype characterized by the absence of therapeutic targets. It shows rapid progression, higher relapse, and poor prognosis, so the establishment of an effective therapeutic target is required. We focused on interleukin-34 (IL-34) that is a novel cytokine relating to inflammation and tumorigenesis. It has been reported that IL-34 correlates with poor prognosis of various cancers. In this study, we evaluated the relationship of IL-34 and prognosis in TNBC using human clinical information and mice model. We found that IL-34 was highly expressed in TNBC, and the survival rate in TNBC was significantly lower in patients with high IL-34 expression. Furthermore, multivariate analysis revealed that IL-34 independently affects prognosis. In murine TNBC model, IL-34 deficiency in tumor cells decreased in vivo tumor growth and increased inflammatory cytokine production from macrophages. These results suggest that tumor-derived IL-34 creates a favorable environment for TNBC cells. Thus, we showed a novel pathological role of IL-34 in TNBC and the potential of IL-34 as a therapeutic target for it.


Asunto(s)
Carcinoma Ductal de Mama/mortalidad , Interleucinas/metabolismo , Neoplasias de la Mama Triple Negativas/mortalidad , Animales , Carcinogénesis/inmunología , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/inmunología , Carcinoma Ductal de Mama/patología , Línea Celular Tumoral , Proliferación Celular , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/inmunología , Técnicas de Inactivación de Genes , Humanos , Interleucinas/genética , Interleucinas/inmunología , Ratones , Pronóstico , Tasa de Supervivencia , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...