Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
BMC Cancer ; 19(1): 924, 2019 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-31521127

RESUMEN

BACKGROUND: Current therapies fail to cure over a third of osteosarcoma patients and around three quarters of those with metastatic disease. "Smac mimetics" (also known as "IAP antagonists") are a new class of anti-cancer agents. Previous work revealed that cells from murine osteosarcomas were efficiently sensitized by physiologically achievable concentrations of some Smac mimetics (including GDC-0152 and LCL161) to killing by the inflammatory cytokine TNFα in vitro, but survived exposure to Smac mimetics as sole agents. METHODS: Nude mice were subcutaneously or intramuscularly implanted with luciferase-expressing murine 1029H or human KRIB osteosarcoma cells. The impacts of treatment with GDC-0152, LCL161 and/or doxorubicin were assessed by caliper measurements, bioluminescence, 18FDG-PET and MRI imaging, and by weighing resected tumors at the experimental endpoint. Metastatic burden was examined by quantitative PCR, through amplification of a region of the luciferase gene from lung DNA. ATP levels in treated and untreated osteosarcoma cells were compared to assess in vitro sensitivity. Immunophenotyping of cells within treated and untreated tumors was performed by flow cytometry, and TNFα levels in blood and tumors were measured using cytokine bead arrays. RESULTS: Treatment with GDC-0152 or LCL161 suppressed the growth of subcutaneously or intramuscularly implanted osteosarcomas. In both models, co-treatment with doxorubicin and Smac mimetics impeded average osteosarcoma growth to a greater extent than either drug alone, although these differences were not statistically significant. Co-treatments were also more toxic. Co-treatment with LCL161 and doxorubicin was particularly effective in the KRIB intramuscular model, impeding primary tumor growth and delaying or preventing metastasis. Although the Smac mimetics were effective in vivo, in vitro they only efficiently killed osteosarcoma cells when TNFα was supplied. Implanted tumors contained high levels of TNFα, produced by infiltrating immune cells. Spontaneous osteosarcomas that arose in genetically-engineered immunocompetent mice also contained abundant TNFα. CONCLUSIONS: These data imply that Smac mimetics can cooperate with TNFα secreted by tumor-associated immune cells to kill osteosarcoma cells in vivo. Smac mimetics may therefore benefit osteosarcoma patients whose tumors contain Smac mimetic-responsive cancer cells and TNFα-producing infiltrating cells.


Asunto(s)
Antineoplásicos/farmacología , Ciclohexanos/farmacología , Pirroles/farmacología , Tiazoles/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Imagen por Resonancia Magnética/métodos , Ratones , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Tomografía de Emisión de Positrones/métodos , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Clin Exp Metastasis ; 38(5): 441-449, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34398333

RESUMEN

Osteosarcoma is the most common form of primary bone cancer and frequently metastasizes to the lungs. Current therapies fail to successfully treat over two thirds of patients with metastatic osteosarcoma, so there is an urgent imperative to develop therapies that effectively target established metastases. Smac mimetics are drugs that work by inhibiting the pro-survival activity of IAP proteins such as cIAP1 and cIAP2, which can be overexpressed in osteosarcomas. In vitro, osteosarcoma cells are sensitive to a range of Smac mimetics in combination with TNFα. This sensitivity has also been demonstrated in vivo using the Smac mimetic LCL161, which inhibited the growth of subcutaneous and intramuscular osteosarcomas. Here, we evaluated the efficacy of LCL161 using mice bearing osteosarcoma metastases without the presence of a primary tumor, modeling the scenario in which a patient's primary tumor had been surgically removed. We demonstrated the ability of LCL161 as a single agent and in combination with doxorubicin to inhibit the growth of, and in some cases eliminate, established pulmonary osteosarcoma metastases in vivo. Resected lung metastases from treated and untreated mice remained sensitive to LCL161 in combination with TNFα ex vivo. This suggested that there was little to no acquired resistance to LCL161 treatment in surviving osteosarcoma cells and implied that tumor microenvironmental factors underlie the observed variation in responses to LCL161.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Osteosarcoma/secundario , Tiazoles/uso terapéutico , Animales , Neoplasias Óseas/patología , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos BALB C , Osteosarcoma/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Nat Commun ; 12(1): 3950, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34168137

RESUMEN

The concept that extracellular vesicles (EVs) from the diet can be absorbed by the intestinal tract of the consuming organism, be bioavailable in various organs, and in-turn exert phenotypic changes is highly debatable. Here, we isolate EVs from both raw and commercial bovine milk and characterize them by electron microscopy, nanoparticle tracking analysis, western blotting, quantitative proteomics and small RNA sequencing analysis. Orally administered bovine milk-derived EVs survive the harsh degrading conditions of the gut, in mice, and is subsequently detected in multiple organs. Milk-derived EVs orally administered to mice implanted with colorectal and breast cancer cells reduce the primary tumor burden. Intriguingly, despite the reduction in primary tumor growth, milk-derived EVs accelerate metastasis in breast and pancreatic cancer mouse models. Proteomic and biochemical analysis reveal the induction of senescence and epithelial-to-mesenchymal transition in cancer cells upon treatment with milk-derived EVs. Timing of EV administration is critical as oral administration after resection of the primary tumor reverses the pro-metastatic effects of milk-derived EVs in breast cancer models. Taken together, our study provides context-based and opposing roles of milk-derived EVs as metastasis inducers and suppressors.


Asunto(s)
Vesículas Extracelulares , Leche/citología , Neoplasias Experimentales/patología , Administración Oral , Animales , Disponibilidad Biológica , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Bovinos , Línea Celular Tumoral , Proliferación Celular , Transición Epitelial-Mesenquimal , Vesículas Extracelulares/química , Vesículas Extracelulares/genética , Femenino , Humanos , Neoplasias Hepáticas Experimentales/patología , Neoplasias Hepáticas Experimentales/secundario , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones Endogámicos BALB C , Neoplasias Experimentales/terapia , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Adolesc Young Adult Oncol ; 9(6): 667-671, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32397787

RESUMEN

Two thirds of metastatic osteosarcoma patients die within 5 years of diagnosis. Improved experimental models of osteosarcoma metastasis will facilitate the development of more effective therapies. Intravenous cancer cell injection can produce lung metastases in nude mice, but this "experimental metastasis" technique has been predominantly applied to a single osteosarcoma cell line (143B) and required injection of 1-2 million cells. Using two human osteosarcoma cell lines, we discovered that transient Natural Killer cell depletion dramatically enhanced the efficiency of experimental pulmonary osteosarcoma metastasis. This technique for modeling osteosarcoma metastasis may enable the identification of better treatments for this aggressive cancer.


Asunto(s)
Células Asesinas Naturales/metabolismo , Neoplasias Pulmonares/secundario , Osteosarcoma/terapia , Administración Intravenosa , Animales , Femenino , Ratones , Ratones Desnudos
5.
Cancers (Basel) ; 12(5)2020 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-32403415

RESUMEN

Osteosarcoma is the most common form of primary bone cancer. Over 20% of osteosarcoma patients present with pulmonary metastases at diagnosis, and nearly 70% of these patients fail to respond to treatment. Previous work revealed that human and canine osteosarcoma cell lines are extremely sensitive to the therapeutic proteasome inhibitor bortezomib in vitro. However, bortezomib has proven disappointingly ineffective against solid tumors including sarcomas in animal experiments and clinical trials. Poor tumor penetration has been speculated to account for the inconsistency between in vitro and in vivo responses of solid tumors to bortezomib. Here we show that the second-generation proteasome inhibitor ixazomib, which reportedly has enhanced solid tumor penetration compared to bortezomib, is toxic to human and canine osteosarcoma cells in vitro. We used experimental osteosarcoma metastasis models to compare the efficacies of ixazomib and bortezomib against primary tumors and metastases derived from luciferase-expressing KRIB or 143B human osteosarcoma cell lines in athymic mice. Neither proteasome inhibitor reduced the growth of primary intramuscular KRIB tumors, however both drugs inhibited the growth of established pulmonary metastases created via intravenous inoculation with KRIB cells, which were significantly better vascularized than the primary tumors. Only ixazomib slowed metastases from KRIB primary tumors and inhibited the growth of 143B pulmonary and abdominal metastases, significantly enhancing the survival of mice intravenously injected with 143B cells. Taken together, these results suggest ixazomib exerts better single agent activity against osteosarcoma metastases than bortezomib. These data provide hope that incorporation of ixazomib, or other proteasome inhibitors that penetrate efficiently into solid tumors, into current regimens may improve outcomes for patients diagnosed with metastatic osteosarcoma.

6.
Data Brief ; 6: 710-4, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26958630

RESUMEN

Unfortunately, the mutagenic activities of chemotherapy and radiotherapy can provoke development of therapy-induced malignancies in cancer survivors. Non-mutagenic anti-cancer therapies may be less likely to trigger subsequent malignant neoplasms. Here we present data regarding the DNA damaging and mutagenic potential of two drugs that antagonize proteins within the Bcl-2 family: ABT-263/Navitoclax and TW-37. Our data reveal that concentrations of these agents that stimulated Bax/Bak-dependent signaling provoked little DNA damage and failed to trigger mutations in surviving cells. The data supplied in this article is related to the research work entitled "Inhibition of Bcl-2 or IAP proteins does not provoke mutations in surviving cells" [1].

7.
Mutat Res ; 787: 15-31, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26943263

RESUMEN

When chemotherapy and radiotherapy are effective, they function by inducing DNA damage in cancerous cells, which respond by undergoing apoptosis. Some adverse effects can result from collateral destruction of non-cancerous cells, via the same mechanism. Therapy-related cancers, a particularly serious adverse effect of anti-cancer treatments, develop due to oncogenic mutations created in non-cancerous cells by the DNA damaging therapies used to eliminate the original cancer. Physiologically achievable concentrations of direct apoptosis inducing anti-cancer drugs that target Bcl-2 and IAP proteins possess negligible mutagenic activity, however death receptor agonists like TRAIL/Apo2L can provoke mutations in surviving cells, probably via caspase-mediated activation of the nuclease CAD. In this study we compared the types of mutations sustained in the HPRT and TK1 loci of clonogenically competent cells following treatment with TRAIL or the alkylating agent ethyl methanesulfonate (EMS). As expected, the loss-of-function mutations in the HPRT or TK1 loci triggered by exposure to EMS were almost all transitions. In contrast, only a minority of the mutations identified in TRAIL-treated clones lacking HPRT or TK1 activity were substitutions. Almost three quarters of the TRAIL-induced mutations were partial or complete deletions of the HPRT or TK1 genes, consistent with sub-lethal TRAIL treatment provoking double strand breaks, which may be mis-repaired by non-homologous end joining (NHEJ). Mis-repair of double-strand breaks following exposure to chemotherapy drugs has been implicated in the pathogenesis of therapy-related cancers. These data suggest that TRAIL too may provoke oncogenic damage to the genomes of surviving cells.


Asunto(s)
Antineoplásicos/toxicidad , Hipoxantina Fosforribosiltransferasa/genética , Mutágenos/toxicidad , Ligando Inductor de Apoptosis Relacionado con TNF/toxicidad , Timidina Quinasa/genética , Secuencia de Bases , Línea Celular , Análisis Mutacional de ADN , Sitios Genéticos , Humanos , Datos de Secuencia Molecular , Eliminación de Secuencia
8.
Oncotarget ; 7(23): 33866-86, 2016 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-27129149

RESUMEN

Outcomes for patients diagnosed with the bone cancer osteosarcoma have not improved significantly in the last four decades. Only around 60% of patients and about a quarter of those with metastatic disease survive for more than five years. Although DNA-damaging chemotherapy drugs can be effective, they can provoke serious or fatal adverse effects including cardiotoxicity and therapy-related cancers. Better and safer treatments are therefore needed. We investigated the anti-osteosarcoma activity of IAP antagonists (also known as Smac mimetics) using cells from primary and metastatic osteosarcomas that arose spontaneously in mice engineered to lack p53 and Rb expression in osteoblast-derived cells. The IAP antagonists SM-164, GDC-0152 and LCL161, which efficiently target XIAP and cIAPs, sensitized cells from most osteosarcomas to killing by low levels of TNFα but not TRAIL. RIPK1 expression levels and activity correlated with sensitivity. RIPK3 levels varied considerably between tumors and RIPK3 was not required for IAP antagonism to sensitize osteosarcoma cells to TNFα. IAP antagonists, including SM-164, lacked mutagenic activity. These data suggest that drugs targeting XIAP and cIAP1/2 may be effective for osteosarcoma patients whose tumors express abundant RIPK1 and contain high levels of TNFα, and would be unlikely to provoke therapy-induced cancers in osteosarcoma survivors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteína 3 que Contiene Repeticiones IAP de Baculovirus/antagonistas & inhibidores , Neoplasias Óseas/tratamiento farmacológico , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Supervivencia Celular/efectos de los fármacos , Ciclohexanos/farmacología , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Osteosarcoma/tratamiento farmacológico , Pirroles/farmacología , Tiazoles/farmacología , Triazoles/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Animales , Proteína 3 que Contiene Repeticiones IAP de Baculovirus/metabolismo , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Compuestos Bicíclicos Heterocíclicos con Puentes/toxicidad , Línea Celular Tumoral , Ciclohexanos/toxicidad , Relación Dosis-Respuesta a Droga , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ratones Noqueados , Osteosarcoma/genética , Osteosarcoma/metabolismo , Osteosarcoma/secundario , Fenotipo , Pirroles/toxicidad , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína de Retinoblastoma/deficiencia , Proteína de Retinoblastoma/genética , Transducción de Señal/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Tiazoles/toxicidad , Transfección , Triazoles/toxicidad , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/metabolismo
9.
Mutat Res ; 777: 23-32, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25916945

RESUMEN

Chemotherapy and radiotherapy can cause permanent damage to the genomes of surviving cells, provoking severe side effects such as second malignancies in some cancer survivors. Drugs that mimic the activity of death ligands, or antagonise pro-survival proteins of the Bcl-2 or IAP families have yielded encouraging results in animal experiments and early phase clinical trials. Because these agents directly engage apoptosis pathways, rather than damaging DNA to indirectly provoke tumour cell death, we reasoned that they may offer another important advantage over conventional therapies: minimisation or elimination of side effects such as second cancers that result from mutation of surviving normal cells. Disappointingly, however, we previously found that concentrations of death receptor agonists like TRAIL that would be present in vivo in clinical settings provoked DNA damage in surviving cells. In this study, we used cell line model systems to investigate the mutagenic capacity of drugs from two other classes of direct apoptosis-inducing agents: the BH3-mimetic ABT-737 and the IAP antagonists LCL161 and AT-406. Encouragingly, our data suggest that IAP antagonists possess negligible genotoxic activity. Doses of ABT-737 that were required to damage DNA stimulated Bax/Bak-independent signalling and exceeded concentrations detected in the plasma of animals treated with this drug. These findings provide hope that cancer patients treated by BH3-mimetics or IAP antagonists may avoid mutation-related illnesses that afflict some cancer survivors treated with conventional DNA-damaging anti-cancer therapies.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Proteínas Inhibidoras de la Apoptosis/genética , Mutación , Proteína X Asociada a bcl-2/genética , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Azocinas/farmacología , Compuestos de Bencidrilo/farmacología , Compuestos de Bifenilo/farmacología , Línea Celular Tumoral , Ensayo Cometa , Daño del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ratones , Mutágenos/farmacología , Nitrofenoles/farmacología , Piperazinas/farmacología , Sulfonamidas/farmacología , Tiazoles/farmacología , Proteína X Asociada a bcl-2/antagonistas & inhibidores , Proteína X Asociada a bcl-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA