Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Future Oncol ; 20(18): 1237-1250, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38592002

RESUMEN

Trastuzumab deruxtecan (T-DXd) is approved for the treatment of human epidermal growth factor receptor 2 (HER2)-low metastatic breast cancer (mBC). Results on T-DXd treatment in HER2-low mBC have so far been limited to clinical trials. DESTINY-Breast Respond HER2-low Europe (NCT05945732) is a multi-center, multi-country, observational, prospective, non-interventional study planning to enroll 1350 patients from 216 sites receiving T-DXd or conventional chemotherapy as their routine clinical care for advanced stage breast cancer in 12 European countries. This non-interventional study will provide real-world insight into T-DXd treatment for HER2-low mBC with data on effectiveness, safety and tolerability, patient-reported outcomes, treatment patterns, geriatric health status and HER2 testing. This will be beneficial for improving guidance to maximize patient treatment benefit.


Trastuzumab deruxtecan (T-DXd; Enhertu®) is a medicine approved to treat cancers that produce a protein called HER2 on the surface of cancer cells. T-DXd works by targeting the HER2 protein to deliver chemotherapy directly to cancer cells. Until recently, breast cancers were classified as HER2-positive (high level of HER2 protein on cancer cells) or HER2-negative (very low level/no HER2 protein on cancer cells). T-DXd was approved for treating patients with HER2-positive advanced breast cancer in Europe in 2022. In 2023 the DESTINY-Breast04 clinical trial showed that T-DXd was more effective than current standard chemotherapies, when treating advanced breast cancer patients with low levels of the HER2 protein (historically classified as HER2-negative cancer). This trial led to the approval of T-DXd for treating advanced HER2-low breast cancer, providing a new treatment option for 50­60% of breast cancer patients. More information is needed about T-DXd treatment in the real world (for patients treated in the hospital, rather than in a clinical trial). This article describes the purpose and design of the DESTINY-Breast Respond HER2-low Europe study, which will collect and report more information about how effective T-DXd treatment is in the real world. This is a large study aiming to include 1350 eligible patients from 12 countries across Europe. Patients will report their experience of side effects (such as nausea and vomiting) to improve management of T-DXd treatment and maximize patient benefit. The study will also examine how elderly patients respond to T-DXd treatment, and how HER2 levels are being tested.Clinical Trial Registration: ICH CGP: NCT05945732, registered on 6 July 2023 (ClinicalTrials.gov).


Asunto(s)
Neoplasias de la Mama , Receptor ErbB-2 , Trastuzumab , Humanos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Femenino , Receptor ErbB-2/metabolismo , Receptor ErbB-2/antagonistas & inhibidores , Trastuzumab/uso terapéutico , Europa (Continente) , Estudios Prospectivos , Inmunoconjugados/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Resultado del Tratamiento , Proyectos de Investigación , Estadificación de Neoplasias , Camptotecina/análogos & derivados
2.
Bioorg Med Chem ; 26(9): 2428-2436, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29673714

RESUMEN

Voltage-gated sodium channels (VGSC) are a well-established drug target for anti-epileptic, anti-arrhythmic and pain medications due to their presence and the important roles that they play in excitable cells. Recently, their presence has been recognized in non-excitable cells such as cancer cells and their overexpression has been shown to be associated with metastatic behavior in a variety of human cancers. The neonatal isoform of the VGSC subtype, Nav1.5 (nNav1.5) is overexpressed in the highly aggressive human breast cancer cell line, MDA-MB-231. The activity of nNav1.5 is known to promote the breast cancer cell invasion in vitro and metastasis in vivo, and its expression in primary mammary tumors has been associated with metastasis and patient death. Metastasis development is responsible for the high mortality of breast cancer and currently there is no treatment available to specifically prevent or inhibit breast cancer metastasis. In the present study, a 3D-QSAR model is used to assist the development of low micromolar small molecule VGSC blockers. Using this model, we have designed, synthesized and evaluated five small molecule compounds as blockers of nNav1.5-dependent inward currents in whole-cell patch-clamp experiments in MDA-MB-231 cells. The most active compound identified from these studies blocked sodium currents by 34.9 ±â€¯6.6% at 1 µM. This compound also inhibited the invasion of MDA-MB-231 cells by 30.3 ±â€¯4.5% at 1 µM concentration without affecting the cell viability. The potent small molecule compounds presented here have the potential to be developed as drugs for breast cancer metastasis treatment.


Asunto(s)
Antineoplásicos/farmacología , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Invasividad Neoplásica/prevención & control , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Diseño de Fármacos , Humanos , Metástasis de la Neoplasia/prevención & control , Relación Estructura-Actividad Cuantitativa , Bloqueadores del Canal de Sodio Activado por Voltaje/síntesis química , Bloqueadores del Canal de Sodio Activado por Voltaje/química
3.
Breast Cancer Res Treat ; 155(2): 261-71, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26780557

RESUMEN

Toll-like receptor 9 (TLR9) is a cellular DNA-receptor widely expressed in cancers. We previously showed that synthetic and self-derived DNA fragments induce TLR9-mediated breast cancer cell invasion in vitro. We investigated here the invasive effects of two nuclease-resistant DNA fragments, a 9-mer hairpin, and a G-quadruplex DNA based on the human telomere sequence, both having native phosphodiester backbone. Cellular uptake of DNAs was investigated with immunofluorescence, invasion was studied with Matrigel-assays, and mRNA and protein expression were studied with qPCR and Western blotting and protease activity with zymograms. TLR9 expression was suppressed through siRNA. Although both DNAs induced TLR9-mediated changes in pro-invasive mRNA expression, only the telomeric G-quadruplex DNA significantly increased cellular invasion. This was inhibited with GM6001 and aprotinin, suggesting MMP- and serine protease mediation. Furthermore, complexing with LL-37, a cathelicidin-peptide present in breast cancers, increased 9-mer hairpin and G-quadruplex DNA uptake into the cancer cells. However, DNA/LL-37 complexes decreased invasion, as compared with DNA-treatment alone. Invasion studies were conducted also with DNA fragments isolated from neoadjuvant chemotherapy-treated breast tumors. Also such DNA induced breast cancer cell invasion in vitro. As with the synthetic DNAs, this invasive effect was reduced by complexing the neoadjuvant tumor-derived DNAs with LL-37. We conclude that 9-mer hairpin and G-quadruplex DNA fragments are nuclease-resistant DNA structures that can act as invasion-inducing TLR9 ligands. Their cellular uptake and the invasive effects are regulated via LL-37. Although such structures may be present in chemotherapy-treated tumors, the clinical significance of this finding requires further studying.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Invasividad Neoplásica/genética , Telómero/genética , Receptor Toll-Like 9/genética , Línea Celular Tumoral , Fragmentación del ADN , ADN de Neoplasias/genética , Femenino , G-Cuádruplex , Humanos , Ligandos , Invasividad Neoplásica/patología , ARN Mensajero/genética , Catelicidinas
4.
J Oral Pathol Med ; 44(8): 571-7, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25338738

RESUMEN

BACKGROUND: Toll-like receptor 9 (TLR9) is a cellular receptor, which recognizes bacterial and host-derived DNA. Stimulation of TLR9 induces cellular invasion via matrix metalloproteinase 13 (MMP-13). The aim of this study was to evaluate the role of TLR9 in invasion of oral tongue squamous cell carcinoma (OTSCC). METHODS: The effects of TLR9 ligands on oral squamous cell carcinoma cell lines were studied with invasion and migration assays, as well as in a myoma organotypic model. RESULTS: The TLR9 ligand, CpG-ODN, increased invasion and migration in OTSCC lines. These effects were reduced by TLR9 siRNA or inhibition with TLR9 antibodies. Immunohistochemical analysis of tissues from 195 patients with OTSCC revealed that TLR9 was expressed in 181/195 carcinomas. The expression of TLR9 was higher in the malignant cells than in the normal epithelium. High TLR9 expression was associated with high MMP-13 expression and poor differentiation. High TLR9 expression was also identified as an independent predictor of poor prognosis (HR 1.810, 95% CI [1.053-3.112]). CONCLUSION: Toll-like receptor 9 mediates OTSCC invasion and migration in vitro and is an independent prognostic factor of OTSCC. Inhibition of TLR9 may be a novel therapeutic opportunity in oral cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Receptor Toll-Like 9/biosíntesis , Neoplasias de la Lengua/metabolismo , Neoplasias de la Lengua/patología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/biosíntesis , Diferenciación Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Femenino , Humanos , Inmunohistoquímica , Masculino , Metaloproteinasa 13 de la Matriz/biosíntesis , Metaloproteinasa 13 de la Matriz/metabolismo , Persona de Mediana Edad , Invasividad Neoplásica , Oligodesoxirribonucleótidos/farmacología , Pronóstico , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Carcinoma de Células Escamosas de Cabeza y Cuello , Receptor Toll-Like 9/genética
5.
Breast Cancer Res Treat ; 142(3): 477-87, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24212717

RESUMEN

TLR9 is a cellular DNA-receptor, which is widely expressed in breast and other cancers. Although synthetic TLR9-ligands induce cancer cell invasion in vitro, the role of TLR9 in cancer pathophysiology has remained unclear. We show here that living cancer cells uptake DNA from chemotherapy-killed cancer cells. We discovered that such DNA induces TLR9- and cathepsin-mediated invasion in living cancer cells. To study whether this phenomenon contributes to treatment responses, triple-negative, human MDA-MB-231 breast cancer cells stably expressing control, or TLR9 siRNA were inoculated orthotopically into nude mice. The mice were treated with vehicle or doxorubicin. The tumor groups exhibited equal decreases in size in response to doxorubicin. However, while the weights of vehicle-treated mice were similar, mice bearing control siRNA tumors became significantly more cachectic in response to doxorubicin, as compared with similarly treated mice bearing TLR9 siRNA tumors, suggesting a TLR9-mediated inflammation at the site of the tumor. In conclusion, our findings propose that DNA released from chemotherapy-killed cancer cells has significant influence on TLR9-mediated biological effects in living cancer cells. Through these mechanisms, tumor TLR9 expression may affect treatment responses to chemotherapy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , ADN/metabolismo , Inflamación/metabolismo , Receptor Toll-Like 9/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Xenoinjertos , Humanos , Inflamación/genética , Inflamación/inmunología , Ratones , Modelos Biológicos , Invasividad Neoplásica , Interferencia de ARN , Receptor Toll-Like 9/genética , Carga Tumoral
6.
Eur J Pharm Sci ; 190: 106571, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37652236

RESUMEN

BACKGROUND: Tumour associated macrophages (TAMs) are important players in breast tumour progression and metastasis. Clinical and preclinical evidence suggests a role for zoledronate (ZOL) in breast cancer metastasis prevention. Further, zoledronate is able to induce inflammatory activation of monocytes and macrophages, which can be favourable in cancer treatments. The inherent bone tropism of zoledronate limits its availability in soft tissues and tumours. In this study we utilised an orthotopic murine breast cancer model to evaluate the possibility to use liposomes (EMP-LIP) to target zoledronate to tumours to modify TAM activation. METHODS: Triple-negative breast cancer 4T1 cells were inoculated in the 4th mammary fat pad of female Balb/c mice. Animals were divided according to the treatment: vehicle, ZOL, EMP-LIP and liposome encapsulated zoledronate (ZOL-LIP). Treatment was done intravenously (with tumour resection) and intraperitoneally (without tumour resection). Tumour growth was followed by bioluminescence in vivo imaging (IVIS) and calliper measurements. Tumour-infiltrating macrophages were assessed by immunohistochemical and immunofluorescence staining. Protein and RNA expression levels of inflammatory transcription factors and cytokines were measured by Western Blotting and Taqman RT-qPCR. RESULTS: Liposome encapsulated zoledronate (ZOL-LIP) treatment suppressed migration of 4T1 cell in vitro. Tumour growth and expression of the angiogenic marker CD34 were reduced upon both ZOL and ZOL-LIP treatment in vivo. Long-term ZOL-LIP treatment resulted in shift towards M1-type macrophage polarization, increased CD4 T cell infiltration and activation of NF-κB indicating changes in intratumoural inflammation, whereas ZOL treatment showed similar but non-significant trends. Moreover, ZOL-LIP had a lower bisphosphonate accumulation in bone compared to free ZOL. CONCLUSION: Results show that the decreased bisphosphonate accumulation in bone promotes the systemic anti-tumour effect of ZOL-LIP by increasing inflammatory response in TNBC tumours via M1-type macrophage activation.


Asunto(s)
Liposomas , Neoplasias de la Mama Triple Negativas , Humanos , Femenino , Ratones , Animales , Ácido Zoledrónico/farmacología , Liposomas/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Línea Celular Tumoral , Difosfonatos/uso terapéutico , Difosfonatos/farmacología , Macrófagos , Ratones Endogámicos BALB C
7.
Front Immunol ; 14: 1179022, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37533856

RESUMEN

Introduction: Bisphosphonates (BPs) are bone-protecting osteoclast inhibitors, typically used in the treatment of osteoporosis and skeletal complications of malignancies. When given in the adjuvant setting, these drugs may also prevent relapses and prolong overall survival in early breast cancer (EBC), specifically among postmenopausal patients. Because of these findings, adjuvant nitrogen-containing BPs (N-BPs), such as zoledronate (ZOL), are now the standard of care for high-risk EBC patients, but there are no benefit-associated biomarkers, and the efficacy remains low. BPs have been demonstrated to possess anti-tumor activities, but the mechanisms by which they provide the beneficial effects in EBC are not known. Methods: We used stably transfected 4T1 breast cancer cells together with suppression of CD73 (sh-CD73) or control cells (sh-NT). We compared ZOL effects on tumor growth and infiltrating lymphocytes (TILs) into tumors and lung metastases using two mouse models. B cell depletion was performed using anti-CD20 antibody. Results: Sh-CD73 4T1 cells were significantly more sensitive to the growth inhibitory effects of n-BPs in vitro. However, while ZOL-induced growth inhibition was similar between the tumor groups in vivo, ZOL enhanced B and T lymphocyte infiltration into the orthotopic tumors with down-regulated CD73. A similar trend was detected in lung metastases. ZOL-induced tumor growth inhibition was found to be augmented with B cell depletion in sh-NT tumors, but not in sh-CD73 tumors. As an internal control, ZOL effects on bone were similar in mice bearing both tumor groups. Discussion: Taken together, these results indicate that ZOL modifies TILs in breast cancer, both in primary tumors and metastases. Our results further demonstrate that B cells may counteract the growth inhibitory effects of ZOL. However, all ZOL-induced TIL effects may be influenced by immunomodulatory characteristics of the tumor.


Asunto(s)
Neoplasias Pulmonares , Neoplasias de la Mama Triple Negativas , Humanos , Animales , Ratones , Ácido Zoledrónico/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Imidazoles/farmacología , Imidazoles/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Linfocitos T
8.
Breast Cancer Res Treat ; 132(2): 411-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21607583

RESUMEN

Toll-like receptor 9 (TLR9) is a cellular DNA-receptor, which is widely expressed in cancer. Synthetic TLR9-ligands induce cancer cell invasion in vitro, but the role of TLR9 in cancer pathophysiology remains unclear. Increased TLR9 expression has been, however, detected in estrogen receptor negative (ER-) breast cancers. In this study, we investigated the effects of ERα expression and sex steroid hormones on TLR9 expression in human ER+ (MCF-7, T47-D) and ER- (MDA-MB-231) breast cancer cell lines in vitro. We also studied TLR9 mRNA expression in archival breast cancer specimens (n = 12) with qRT-PCR, using primer sets that detect only the TLR9A isoform or the isoforms A and B (TLR9A/B). The TLR9 mRNA expression was detected in 10/12 specimens with both primer sets, and in 1/12 with only the TLR9A or the TLR9A/B primer sets. The basal TLR9 mRNA expression levels were significantly lower in the ER+ cell lines as compared with the ER- MDA-MB-231 cells. The transfection of ERα cDNA into MDA-MB-231 cells also resulted in down-regulation of TLR9 expression. While sex steroids had no effect on TLR9 expression in MCF-7 cells, testosterone (10(-8) M) induced TLR9 expression in MDA-MB-231 and T47-D cells. Although bicalutamide blocked this testosterone effect in MDA-MB-231 cells, in T47-D cells bicalutamide increased TLR9 expression and only partially blocked the testosterone effects. Estradiol (10(-8) M) induced TLR9 expression in T47-D cells. The invasive effects of synthetic TLR9-ligands were augmented by testosterone in vitro. This effect was lost in TLR9 siRNA MDA-MB-231 cells and also decreased by over-expression of ERα, which also inhibited NF-κB activation by TLR9-ligands. In conclusion, expression of TLR9 isoforms A and B can be detected in clinical breast cancer specimens. The ERα and sex steroid hormones regulate TLR9 expression and invasive effects in the breast cancer cells. Also, the commonly used hormonal cancer therapy bicalutamide affects TLR9 expression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular , Receptor alfa de Estrógeno/metabolismo , Hormonas Esteroides Gonadales/metabolismo , Receptor Toll-Like 9/metabolismo , Antagonistas de Andrógenos/farmacología , Anilidas/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Estradiol/metabolismo , Receptor alfa de Estrógeno/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ligandos , FN-kappa B/genética , FN-kappa B/metabolismo , Invasividad Neoplásica , Nitrilos/farmacología , Isoformas de Proteínas , Interferencia de ARN , ARN Mensajero/metabolismo , Transducción de Señal , Testosterona/análogos & derivados , Testosterona/metabolismo , Receptor Toll-Like 9/genética , Compuestos de Tosilo/farmacología , Transfección
9.
Breast Cancer Res Treat ; 135(2): 481-93, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22847512

RESUMEN

Toll-like receptor-9 (TLR9) is a DNA receptor widely expressed in cancers. Although synthetic TLR9 ligands induce cancer cell invasion in vitro, the role of TLR9 in cancer pathophysiology is unclear. We discovered that low tumor TLR9 expression is associated with significantly shortened disease-specific survival in patients with triple negative but not with ER+ breast cancers. A likely mechanism of this clinical finding involves differential responses to hypoxia. Our pre-clinical studies indicate that while TLR9 expression is hypoxia-regulated, low TLR9 expression has different effects on triple negative and ER+ breast cancer invasion in hypoxia. Hypoxia-induced invasion is augmented by TLR9 siRNA in triple negative, but not in ER+ breast cancer cells. This is possibly due to differential TLR9-regulated TIMP-3 expression, which remains detectable in ER+ cells but disappears from triple-negative TLR9 siRNA cells in hypoxia. Our results demonstrate a novel role for this innate immunity receptor in cancer biology and suggest that TLR9 expression may be a novel marker for triple-negative breast cancer patients who are at a high risk of relapse. Furthermore, these results suggest that interventions or events, which induce hypoxia or down-regulate TLR9 expression in triple-negative breast cancer cells may actually induce their spread.


Asunto(s)
Neoplasias de la Mama/metabolismo , Expresión Génica , Receptor Toll-Like 9/metabolismo , Animales , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Hipoxia de la Célula , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Estimación de Kaplan-Meier , Metaloproteinasas de la Matriz Secretadas/genética , Metaloproteinasas de la Matriz Secretadas/metabolismo , Ratones , Ratones Desnudos , Invasividad Neoplásica , Trasplante de Neoplasias , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/genética , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Receptor Toll-Like 9/genética , Carga Tumoral , Regulación hacia Arriba
10.
Oncogenesis ; 11(1): 53, 2022 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-36085291

RESUMEN

The pathophysiology and the optimal treatment of breast neuroendocrine tumours (NETs) are unknown. We compared the mutational profiles of breast NETs (n = 53) with those of 724 publicly available invasive ductal carcinoma (IDC) and 98 pancreatic NET (PNET) cases. The only significantly different pathogenetic or unknown variant rate between breast NETs and IDCs was detected in the TP53 (11.3% in breast NETs and 41% in IDCs, adjusted p value 0.027) and ADCK2 (9.4% in breast NETs vs. 0.28% in IDCs, adjusted p value 0.045) genes. Between breast NETs and PNETs, different pathogenetic or unknown variant frequencies were detected in 30 genes. For example, MEN1 was mutated in only 6% of breast NETs and 37% in PNETs (adjusted p value 0.00050), and GATA3 pathogenetic or unknown variants were only found in 17.0% of breast NETs and 0% in PNETs (adjusted p value 0.0010). The most commonly affected oncogenic pathways in the breast NET cases were PI3K/Akt/mTOR, NOTCH and RTK-RAS pathways. Breast NETs had typically clock-like mutational signatures and signatures associated with defective DNA mismatch repair in their mutational landscape. Our results suggest that the breast NET mutational profile more closely resembles that of IDCs than that of PNETs. These results also revealed several potentially druggable targets, such as MMRd, in breast NETs. In conclusion, breast NETs are indeed a separate breast cancer entity, but their optimal treatment remains to be elucidated.

11.
Histopathology ; 59(4): 643-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22014045

RESUMEN

AIMS: Toll-like receptor 9 (TLR-9) is a cellular DNA receptor that has been linked previously to invasion in various cancers. The aim of this study was to investigate TLR-9 expression and its possible association with prognosis in oesophageal adenocarcinoma. METHODS AND RESULTS: Immunohistochemical TLR-9 expression was graded in clinical specimens (n = 76) of oesophageal adenocarcinoma. The TLR-9 immunostaining intensity was compared with tumour grade, stage and indicators of proliferation, apoptosis and tumour vascular supply. High TLR-9 expression correlated with advanced tumour stage, tumour unresectability, poor differentiation and high proliferation. Strong immunoreactivity of TLR-9 also indicated poor overall survival. CONCLUSIONS: High TLR-9 expression is associated with poor differentiation, a high proliferation rate and disseminated disease. Accordingly, increased TLR-9 expression may contribute to the growth and metastatic properties of oesophageal adenocarcinoma.


Asunto(s)
Adenocarcinoma/metabolismo , Biomarcadores de Tumor/análisis , Neoplasias Esofágicas/metabolismo , Receptor Toll-Like 9/biosíntesis , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Adulto , Anciano , Apoptosis/fisiología , Biomarcadores de Tumor/metabolismo , Diferenciación Celular , Proliferación Celular , Neoplasias Esofágicas/mortalidad , Neoplasias Esofágicas/patología , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Modelos de Riesgos Proporcionales , Receptor Toll-Like 9/análisis
12.
Sci Rep ; 11(1): 6035, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33727591

RESUMEN

CD73 is a cell surface ecto-5'-nucleotidase, which converts extracellular adenosine monophosphate to adenosine. High tumor CD73 expression is associated with poor outcome among triple-negative breast cancer (TNBC) patients. Here we investigated the mechanisms by which CD73 might contribute to TNBC progression. This was done by inhibiting CD73 with adenosine 5'-(α, ß-methylene) diphosphate (APCP) in MDA-MB-231 or 4T1 TNBC cells or through shRNA-silencing (sh-CD73). Effects of such inhibition on cell behavior was then studied in normoxia and hypoxia in vitro and in an orthotopic mouse model in vivo. CD73 inhibition, through shRNA or APCP significantly decreased cellular viability and migration in normoxia. Inhibition of CD73 also resulted in suppression of hypoxia-induced increase in viability and prevented cell protrusion elongation in both normoxia and hypoxia in cancer cells. Sh-CD73 4T1 cells formed significantly smaller and less invasive 3D organoids in vitro, and significantly smaller orthotopic tumors and less lung metastases than control shRNA cells in vivo. CD73 suppression increased E-cadherin and decreased vimentin expression in vitro and in vivo, proposing maintenance of a more epithelial phenotype. In conclusion, our results suggest that CD73 may promote early steps of tumor progression, possibly through facilitating epithelial-mesenchymal transition.


Asunto(s)
5'-Nucleotidasa/metabolismo , Transición Epitelial-Mesenquimal , Neoplasias Pulmonares/enzimología , Neoplasias Mamarias Animales/enzimología , Proteínas de Neoplasias/metabolismo , Neoplasias de la Mama Triple Negativas/enzimología , 5'-Nucleotidasa/genética , Animales , Línea Celular Tumoral , Femenino , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
13.
Prostate ; 70(8): 817-24, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20054821

RESUMEN

BACKGROUND: Toll-like receptor-9 (TLR9) is a cellular receptor for bacterial and vertebrate DNA. In addition to cells of the immune system, it is also expressed in various human cancer cell lines, including prostate cancer. We demonstrated previously that synthetic TLR9 ligands induce matrix metalloproteinase-13-mediated invasion in TLR9-expressing prostate cancer cells in vitro. Other studies have suggested possible sex steroid regulation of the function of the various TLRs. The role of TLR9 in the pathophysiology of prostate or any cancer is, however, unknown. METHODS: Expression of TLR9, androgen receptor (AR), or the estrogen receptors alpha (ERalpha) and beta (ERbeta) were studied with immunohistochemistry in prostate cancer (n = 62) and benign prostatic hyperplasia (n = 45) specimens. TLR9 staining scores were compared with tumor stage, Gleason score, prostate-specific antigen (PSA) concentrations before tissue sampling and with the staining scores of AR, ERalpha, and ERbeta. RESULTS: TLR9 expression was statistically significantly increased in prostate cancer epithelium and stroma, as compared with the same cellular compartments in benign hyperplasia. Significantly increased (P = 0.04) TLR9 expression was detected in cancers with high Gleason score (>7, n = 23), as compared with lower Gleason scores (< or =7, n = 39). No statistically significant associations were detected between TLR9 expression scores and PSA concentrations or tumor staging. Prostate adenocarcinoma cells were all positive for TLR9, AR, and ERbeta but negative for ERalpha expression. In cancer stroma cells, increased TLR9 expression was associated with increased ERalpha expression. CONCLUSIONS: Expression of TLR9 is increased in prostate cancer specimens, especially in the most poorly differentiated forms.


Asunto(s)
Adenocarcinoma/metabolismo , Próstata/metabolismo , Hiperplasia Prostática/metabolismo , Neoplasias de la Próstata/metabolismo , Receptor Toll-Like 9/metabolismo , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Epitelio/metabolismo , Epitelio/patología , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Próstata/patología , Antígeno Prostático Específico/metabolismo , Hiperplasia Prostática/patología , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo
14.
Prostate ; 69(6): 652-61, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19152406

RESUMEN

BACKGROUND: Macrophage inhibitory cytokine-1 (MIC-1) belongs to the bone morphogenic protein/transforming growth factor-beta (BMP/TGF-beta) superfamily. Serum MIC-1 concentrations are elevated in patients with advanced prostate cancer. The effects of MIC-1 on prostate cancer bone metastases are unknown. METHODS: In vitro effects of MIC-1 on osteoblast differentiation and activity were analyzed with alkaline phosphatase and mineralization assays; osteoclast numbers were counted microscopically. MIC-1 effects on TLR9 expression were studied with Western blotting. Human Du-145 prostate cancer cells were stably transfected with a cDNA encoding for mature MIC-1 or with an empty vector. The in vivo growth characteristics of the characterized cells were studied with the intra-tibial model of bone metastasis. Tumor associated bone changes were viewed with X-rays, histology, and histomorphometry. Bone formation was assayed by measuring serum PINP. RESULTS: MIC-1 induced osteoblast differentiation and activity and osteoclast formation in vitro. These effects were independent of TLR9 expression, which was promoted by MIC-1. Both MIC-1 and control tumors induced mixed sclerotic/lytic bone lesions, but MIC-1 increased the osteolytic component of tumors. Osteoclast formation at the tumor-bone interface was significantly higher in the MIC-1 tumors, whereas bone formation was significantly higher in the control mice. At sacrifice, the mice bearing MIC-1 tumors were significantly lighter with significantly smaller tumors. CONCLUSIONS: MIC-1 up-regulates TLR9 expression in various cells. MIC-1 stimulates both osteoblast and osteoclast differentiation in vitro, independently of TLR9. MIC-1 over-expressing prostate cancer cells that grow in bone induce osteoclast formation and cachexia.


Asunto(s)
Neoplasias Óseas/secundario , Regulación Neoplásica de la Expresión Génica , Factor 15 de Diferenciación de Crecimiento/genética , Metástasis de la Neoplasia/genética , Osteoclastos/fisiología , Neoplasias de la Próstata/patología , Pérdida de Peso/genética , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Noqueados , Osteoblastos/fisiología , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética
15.
Mol Cancer Res ; 6(10): 1534-43, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18922969

RESUMEN

Toll-like receptor 9 (TLR9) belongs to the innate immune system and recognizes microbial and vertebrate DNA. We showed previously that treatment with the TLR9-agonistic ODN M362 (a CpG sequence containing oligonucleotide) induces matrix metalloproteinase-13-mediated invasion in TLR9-expressing human cancer cell lines. Here, we further characterized the role of the TLR9 pathway in this process. We show that CpG oligonucleotides induce invasion in macrophages from wild-type C57/B6 and MyD88 knockout mice and in human MDA-MB-231 breast cancer cells lacking MyD88 expression. This effect was significantly inhibited in macrophages from TLR9 knockout mice and in human MDA-MB-231 breast cancer cells stably expressing TLR9 small interfering RNA or dominant-negative tumor necrosis factor receptor-associated factor 6 (TRAF6). Sequence modifications to the CpG oligonucleotides that targeted the stem loop and other secondary structures were shown to influence the invasion-inducing effect in MDA-MB-231 cells. In contrast, methylation of the cytosine residues of the parent CpG oligonucleotide did not affect the TLR9-mediated invasion compared with the unmethylated parent CpG oligonucleotide. Finally, expression of TLR9 was studied in clinical breast cancer samples and normal breast epithelium with immunohistochemistry. TLR9 staining localized in epithelial cells in both cancer and normal samples. The mean TLR9 staining intensity was significantly increased in the breast cancer cells compared with normal breast epithelial cells. In conclusion, our results suggest that TLR9 expression is increased in breast cancer and CpG oligonucleotide-induced cellular invasion is mediated via TLR9 and TRAF6, independent of MyD88. Further, our findings suggest that the structure and/or stability of DNA may influence the induction of TLR9-mediated invasion in breast cancer.


Asunto(s)
Invasividad Neoplásica/patología , Oligodesoxirribonucleótidos/farmacología , Receptor Toll-Like 9/metabolismo , Animales , Secuencia de Bases , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Metilación de ADN/efectos de los fármacos , Femenino , Genes Dominantes , Humanos , Ratones , Datos de Secuencia Molecular , Conformación de Ácido Nucleico/efectos de los fármacos , Ácidos Nucleicos Heterodúplex/metabolismo , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/genética , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo
16.
Int J Cancer ; 123(5): 1034-42, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18546262

RESUMEN

Most breast cancer metastases in bone form osteolytic lesions, but the mechanisms of tumor-induced bone resorption and destruction are not fully understood. Although it is well recognized that Wnt/beta-catenin signaling is important for breast cancer tumorigenesis, the role of this pathway in breast cancer bone metastasis is unclear. Dickkopf1 (Dkk1) is a secreted Wnt/beta-catenin antagonist. In the present study, we demonstrated that activation of Wnt/beta-catenin signaling enhanced Dkk1 expression in breast cancer cells and that Dkk1 overexpression is a frequent event in breast cancer. We also found that human breast cancer cell lines that preferentially form osteolytic bone metastases exhibited increased levels of Wnt/beta-catenin signaling and Dkk1 expression. Moreover, we showed that breast cancer cell-produced Dkk1 blocked Wnt3A-induced osteoblastic differentiation and osteoprotegerin (OPG) expression of osteoblast precursor C2C12 cells and that these effects could be neutralized by a specific anti-Dkk1 antibody. In addition, we found that breast cancer cell conditioned media were able to block Wnt3A-induced NF-kappaB ligand reduction in C2C12 cells. Finally, we demonstrated that conditioned media from breast cancer cells in which Dkk1 expression had been silenced via RNAi were unable to block Wnt3A-induced C2C12 osteoblastic differentiation and OPG expression. Taken together, these results suggest that breast cancer-produced Dkk1 may be an important mechanistic link between primary breast tumors and secondary osteolytic bone metastases.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Osteoblastos/patología , Osteólisis , Osteoprotegerina/metabolismo , Neoplasias Óseas/metabolismo , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , FN-kappa B/metabolismo , Osteoblastos/metabolismo , Ligando RANK/metabolismo , Regulación hacia Arriba , Proteínas Wnt/metabolismo , Proteína Wnt3 , Proteína Wnt3A , beta Catenina/metabolismo
17.
Cancer Epidemiol Biomarkers Prev ; 16(3): 532-7, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17372249

RESUMEN

Macrophage-inhibitory cytokine-1 (MIC-1) is a divergent member of the transforming growth factor beta superfamily. It is up-regulated by nonsteroidal anti-inflammatory drugs and is highly expressed in human prostate cancer leading to high serum MIC-1 concentrations with advanced disease. A role for MIC-1 has been implicated in the process of early bone formation, suggesting that it may also mediate sclerosis at the site of prostate cancer bone metastases. Consequently, the aim of this study was to retrospectively determine the relationship of serum MIC-1 concentration and other markers related to current and future prostate cancer bone metastasis in a cohort of 159 patients with prostate cancer. Serum markers included cross-linked carboxy-terminal telopeptide of type I collagen, prostate-specific antigen, and amino-terminal propeptide of type I procollagen (PINP). The mean values of all the biomarkers studied were significantly higher in patients with baseline bone metastases (BM+, n = 35), when compared with those without bone metastases (BM-, n = 124). In a multivariate logistic model, both MIC-1 and PINP independently predicted the presence of baseline bone metastasis. Based on receiver operator curve analysis, the best predictor for the presence of baseline bone metastasis was MIC-1, which was significantly better than carboxy-terminal telopeptide of type I collagen, prostate-specific antigen, and PINP. Patients who experienced bone relapse had significantly higher levels of baseline MIC-1 compared with patients who did not (1476.7 versus 988.4; P = 0.03). Current use of acetylsalicylic acid did not influence serum MIC-1 levels in this cohort. Although requiring validation prospectively, these results suggest that serum MIC-1 determination may be a valuable tool for the diagnosis of current and future bone metastases in patients with prostate cancer.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias Óseas/sangre , Neoplasias Óseas/secundario , Citocinas/sangre , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/patología , Anciano , Anciano de 80 o más Años , Análisis Discriminante , Ensayo de Inmunoadsorción Enzimática , Factor 15 de Diferenciación de Crecimiento , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Antígeno Prostático Específico/sangre , Curva ROC , Estudios Retrospectivos
18.
Mol Cancer Res ; 4(7): 437-47, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16849519

RESUMEN

Toll-like receptor 9 (TLR9) recognizes microbial DNA. We show here that TLR9 protein is expressed in human breast cancer cells and clinical breast cancer samples. Stimulation of TLR9-expressing breast cancer cells with the TLR9 agonistic CpG oligonucleotides (1-10 mumol/L) dramatically increased their in vitro invasion in both Matrigel assays and three-dimensional collagen cultures. Similar effects on invasion were seen in TLR9-expressing astrocytoma and glioblastoma cells and in the immortalized human breast epithelial cell line MCF-10A. This effect was not, however, dependent on the CpG content of the TLR9 ligands because the non-CpG oligonucleotides induced invasion of TLR9-expressing cells. CpG or non-CpG oligonucleotide-induced invasion in MDA-MB-231 cells was blunted by chloroquine and they did not induce invasion of TLR9(-) breast cancer cells. Treatment of MDA-MB-231 cells with CpG or non-CpG oligonucleotides induced the formation of approximately 50-kDa gelatinolytic band in zymograms. This band and the increased invasion were abolished by a matrix metalloproteinase (MMP) inhibitor GM6001 but not by a serine proteinase inhibitor aprotinin. Furthermore, CpG oligonucleotide treatment decreased tissue inhibitor of metalloproteinase-3 expression and increased levels of active MMP-13 in TLR9-expressing but not TLR9(-) breast cancer cells without affecting MMP-8. Neutralizing anti-MMP-13 antibodies inhibited the CpG oligonucleotide-induced invasion. These findings suggest that infections may promote cancer progression through a novel TLR9-mediated mechanism. They also propose a new molecular target for cancer therapy, because TLR9 has not been associated with cancer invasiveness previously.


Asunto(s)
Neoplasias de la Mama/enzimología , Metaloproteinasas de la Matriz/metabolismo , Receptor Toll-Like 9/agonistas , Anticuerpos/farmacología , Astrocitoma/enzimología , Astrocitoma/patología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Colagenasas/inmunología , Colagenasas/metabolismo , Islas de CpG , Proteínas de Unión al ADN/genética , Glioblastoma/enzimología , Glioblastoma/patología , Humanos , Metaloproteinasa 13 de la Matriz , Metaloproteinasa 8 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz , Invasividad Neoplásica , Oligonucleótidos/genética , Oligonucleótidos/farmacología , Receptor Toll-Like 9/biosíntesis , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Transactivadores
19.
Eur J Pharmacol ; 570(1-3): 27-37, 2007 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-17640631

RESUMEN

Bisphosphonates are widely used inhibitors of bone resorption. They also inhibit the growth of various cancer cells in vitro, but the clinical significance of this effect is unclear. The cancer growth inhibitory effects of nitrogen-containing bisphosphonates, (i.e. zoledronate) have been attributed to their ability to inhibit the mevalonate pathway. We have shown that bisphosphonates also induce p38 activation, which signals resistance against the drug-induced growth inhibition through an unknown mechanism. We show here that zoledronate induces a G1/S cell cycle arrest in human MDA-MB-231 breast cancer cells. Furthermore, p38 inhibitor augments bisphosphonate-induced growth inhibition by inducing an additional G2-phase cell cycle arrest. We also show that the nitrogen-containing bisphosphonate-induced effects on p38 phosphorylation occur before accumulation of unprenylated Rap1A or Rac1 activation. Geranylgeranyl pyrophosphate, an end-product of the mevalonate pathway, reversed the accumulation of unprenylated Rap1A but not phosphorylation of p38. Geranylgeranyl pyrophosphate also reversed n-BP induced growth inhibition, but the completeness of this reversal was nitrogen-containing bisphosphonate concentration dependent. Also mevastatin induced the accumulation of unprenylated Rap1A, but it did not induce p38 phosphorylation. In conclusion, our results suggest that in addition to the previously reported effects on apoptosis, nitrogen-containing bisphosphonates also inhibit the growth of MDA-MB-231 breast cancer cells by inducing G1/S cell cycle arrest. The bisphosphonate-induced p38 activation signals for resistance against these drugs, by promoting progression through the G2/M-checkpoint. Of these pathways only growth inhibition is mediated via inhibition of the mevalonate pathway in MDA-MB-231 cells. Combining p38 inhibitors with bisphosphonates may result in increased anti-cancer efficacy.


Asunto(s)
Antineoplásicos/farmacología , Difosfonatos/farmacología , Ácido Mevalónico/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Neoplasias de la Mama , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , ADN/metabolismo , Humanos , Lovastatina/análogos & derivados , Lovastatina/farmacología , Nitrógeno , Fosfatos de Poliisoprenilo/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas de Unión al GTP rap1/metabolismo
20.
Eur J Pharmacol ; 572(2-3): 102-10, 2007 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-17632097

RESUMEN

Nonsteroidal anti-inflammatory drugs (NSAIDs) inhibit cyclooxygenases (COX) and are widely used for post-trauma musculoskeletal analgesia. In animal models, NSAIDs have been reported to delay fracture healing and cause non-union, possibly due to the drug-induced inhibition of osteoblast recruitment and differentiation. To further investigate the cellular effects of these drugs in the context of bone healing, we examined the effects of COX-1 inhibitor indomethacin and COX-2 inhibitors, parecoxib and NS398 on osteoclast and osteoblast differentiation and activity in vitro. We discovered that all tested COX-inhibitors significantly inhibited osteoclast differentiation, by 93%, 94% and 74% of control for 100 microM indomethacin, 100 microM parecoxib and 3 microM NS398, respectively. Furthermore, inhibition of COX-2 reduced also the resorption activity of mature osteoclasts. All tested COX-inhibitors also significantly inhibited osteoblast differentiation from human mesenchymal stem cells. Simultaneously, the number of adipocytes was significantly increased. The adipocyte covered areas in the cultures with 1 microM indomethacin, 1 microM parecoxib and 3 microM NS398 were 9%, 29% and 24%, respectively, as compared with 6% in the control group. This data suggests that COX-2 inhibition disturbs bone remodelling by inhibiting osteoclast differentiation and diverting stem cell differentiation towards adipocyte lineage instead of osteoblast lineage. In conclusion, our results further suggest cautious use of COX-2 inhibitors after osseous trauma.


Asunto(s)
Adipocitos/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/efectos adversos , Células Madre Mesenquimatosas/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Adipocitos/citología , Animales , Bovinos , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Células Cultivadas , Inhibidores de la Ciclooxigenasa 2/efectos adversos , Inhibidores de la Ciclooxigenasa 2/farmacología , Inhibidores de la Ciclooxigenasa/farmacología , Humanos , Técnicas In Vitro , Indometacina/efectos adversos , Indometacina/farmacología , Isoxazoles/efectos adversos , Isoxazoles/farmacología , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Nitrobencenos/efectos adversos , Nitrobencenos/farmacología , Osteoblastos/citología , Osteoclastos/citología , Ratas , Ratas Sprague-Dawley , Sulfonamidas/efectos adversos , Sulfonamidas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA