Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Angiogenesis ; 19(2): 229-44, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26961182

RESUMEN

BACKGROUND: Metronomic chemotherapy has shown promising activity against solid tumors and is believed to act in an antiangiogenic manner. The current study describes and quantifies the therapeutic efficacy, and mode of activity, of metronomic gemcitabine and a dedicated antiangiogenic agent (DC101) in patient-derived xenografts of pancreatic cancer. METHODS: Two primary human pancreatic cancer xenograft lines were dosed metronomically with gemcitabine or DC101 weekly. Changes in tumor growth, vascular function, and metabolism over time were measured with magnetic resonance imaging, positron emission tomography, and immunofluorescence microscopy to determine the anti-tumor effects of the respective treatments. RESULTS: Tumors treated with metronomic gemcitabine were 10-fold smaller than those in the control and DC101 groups. Metronomic gemcitabine, but not DC101, reduced the tumors' avidity for glucose, proliferation, and apoptosis. Metronomic gemcitabine-treated tumors had higher perfusion rates and uniformly distributed blood flow within the tumor, whereas perfusion rates in DC101-treated tumors were lower and confined to the periphery. DC101 treatment reduced the tumor's vascular density, but did not change their function. In contrast, metronomic gemcitabine increased vessel density, improved tumor perfusion transiently, and decreased hypoxia. CONCLUSION: The aggregate data suggest that metronomic gemcitabine treatment affects both tumor vasculature and tumor cells continuously, and the overall effect is to significantly slow tumor growth. The observed increase in tumor perfusion induced by metronomic gemcitabine may be used as a therapeutic window for the administration of a second drug or radiation therapy. Non-invasive imaging could be used to detect early changes in tumor physiology before reductions in tumor volume were evident.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Desoxicitidina/análogos & derivados , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Administración Metronómica , Inhibidores de la Angiogénesis/farmacología , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Proliferación Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Humanos , Masculino , Ratones SCID , Microvasos/efectos de los fármacos , Microvasos/patología , Necrosis , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/patología , Perfusión , Gemcitabina
2.
Proc Natl Acad Sci U S A ; 109(18): 6993-8, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22509029

RESUMEN

Vascular smooth muscle cells (VSMC) have been suggested to arise from various developmental sources during embryogenesis, depending on the vascular bed. However, evidence also points to a common subpopulation of vascular progenitor cells predisposed to VSMC fate in the embryo. In the present study, we use binary transgenic reporter mice to identify a Tie1(+)CD31(dim)vascular endothelial (VE)-cadherin(-)CD45(-) precursor that gives rise to VSMC in vivo in all vascular beds examined. This precursor does not represent a mature endothelial cell, because a VE-cadherin promoter-driven reporter shows no expression in VSMC during murine development. Blockade of Notch signaling in the Tie1(+) precursor cell, but not the VE-cadherin(+) endothelial cell, decreases VSMC investment of developing arteries, leading to localized hemorrhage in the embryo at the time of vascular maturation. However, Notch signaling is not required in the Tie1(+) precursor after establishment of a stable artery. Thus, Notch activity is required in the differentiation of a Tie1(+) local precursor to VSMC in a spatiotemporal fashion across all vascular beds.


Asunto(s)
Diferenciación Celular/fisiología , Mioblastos del Músculo Liso/citología , Mioblastos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Neovascularización Fisiológica , Receptores Notch/metabolismo , Animales , Antígenos CD/genética , Arterias/embriología , Arterias/crecimiento & desarrollo , Arterias/metabolismo , Secuencia de Bases , Cadherinas/deficiencia , Cadherinas/genética , Diferenciación Celular/genética , Cartilla de ADN/genética , Femenino , Antígenos Comunes de Leucocito/deficiencia , Antígenos Comunes de Leucocito/genética , Ratones , Ratones Transgénicos , Neovascularización Fisiológica/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Embarazo , Receptor TIE-1/metabolismo , Receptores Notch/antagonistas & inhibidores , Transducción de Señal
3.
DNA Repair (Amst) ; 139: 103689, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38749239

RESUMEN

The effectiveness of radiotherapy depends on the sensitivities of 'normal' and cancer cells to the administered radiation dose. Increasing the radiosensitivity of cancers by inhibiting DNA damage repair is a goal of much current research, however success depends on avoiding concomitant sensitization of normal tissues inevitably irradiated during therapy. In this study we investigated the mechanisms of radiosensitization for DNA-PK and PARP inhibitors by examining the impacts on proliferating vs quiescent cell populations. Experiments were performed in BRCA1/2null and wild-type parental cancer models in vitro and in vivo. Overall AZD7648 has greater radiosensitizing activity relative to Olaparib, with BRCA2-deficient models showing the greatest sensitivity. However, DNA-PK inhibitor AZD7648 also produced greater toxicity in all irradiated mice. While both DNA-PK and PARP inhibition sensitizes wild type tumor cells to radiation, in BRCA1/2 deficient cells PARP inhibition by Olaparib had limited radiosensitization capacity. Quiescent cells are more radioresistant than proliferating cells, and these were also effectively sensitized by AZD7648 while Olaparib was unable to increase radiation-induced cell kill, even in BRCA1/2null cells. These findings underscore the distinct mechanisms of radiosensitization for DNA-PK and PARP inhibitors. While DNA-PK inhibitors are able to target both proliferating and non-proliferating tumor cells for greater overall anti-cancer benefit, their application is limited by exacerbation of normal tissue toxicities. Conversely, PARP inhibitors exhibit selective activity for proliferating cells, providing a mechanism for targeting activity to cancers, but due to poor activity in non-proliferating cells they have an overall reduced impact on tumor growth control. This study highlights the importance of creating a therapeutic ratio with DNA damage repair inhibition radiation sensitizing strategies.


Asunto(s)
Proteína BRCA1 , Proteína BRCA2 , Proteína Quinasa Activada por ADN , Ftalazinas , Piperazinas , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Fármacos Sensibilizantes a Radiaciones , Ftalazinas/farmacología , Piperazinas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Animales , Humanos , Proteína BRCA1/metabolismo , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Proteína Quinasa Activada por ADN/metabolismo , Ratones , Línea Celular Tumoral , Femenino , Proteína BRCA2/genética , Proliferación Celular/efectos de los fármacos , Tolerancia a Radiación/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Mol Cancer Ther ; 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38781104

RESUMEN

Inhibitors of DNA-PK sensitize cancers to radiotherapy and DNA-damaging chemotherapies, with candidates in clinical trials. However, the degree to which DNA-PK inhibitors also sensitize normal tissues remains poorly characterized. In this study we compare tumor growth control and normal tissue sensitization following DNA-PK inhibitors in combination with radiation and etoposide. FaDu tumor xenografts implanted in mice were treated with 10 - 15Gy irradiation ± 3 - 100 mg/kg AZD7648. A dose-dependent increase in time to tumor volume doubling following AZD7648 was proportional to an increase in toxicity scores of the overlying skin. Similar effects were seen in the intestinal jejunum, tongue and FaDu tumor xenografts of mice assessed for proliferation rates at 3.5 days after treatment with etoposide or 5Gy whole body irradiation ± DNA-PK inhibitors AZD7648 or peposertib (M3814). Additional organs were examined for sensitivity to DNA-PK inhibitor activity in ATM-deficient mice, where DNA-PK activity is indicated by surrogate marker γH2AX. Inhibition was observed in heart, brain, pancreas, thymus, tongue and salivary glands of ATM-deficient mice treated with the DNA-PK inhibitors relative to radiation alone. Similar reductions are also seen in ATM-deficient FaDu tumor xenografts where both pDNA-PK and γH2AX staining could be performed. Conclusions: DNA-PK inhibitor-mediated sensitization to radiation and DNA-damaging chemotherapy is not limited to tumor tissues, but also extends to normal tissues sustaining DNA damage. These data are useful for interpretation of the sensitizing effects of DNA damage repair inhibitors, where a therapeutic index showing greater cell-killing effects on cancer cells is crucial for optimal clinical translation.

5.
Radiother Oncol ; 196: 110326, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38735536

RESUMEN

PURPOSE: The oxygen depletion hypothesis has been proposed as a rationale to explain the observed phenomenon of FLASH-radiotherapy (FLASH-RT) sparing normal tissues while simultaneously maintaining tumor control. In this study we examined the distribution of DNA Damage Response (DDR) markers in irradiated 3D multicellular spheroids to explore the relationship between FLASH-RT protection and radiolytic-oxygen-consumption (ROC) in tissues. METHODS: Studies were performed using a Varian Truebeam linear accelerator delivering 10 MeV electrons with an average dose rate above 50 Gy/s. Irradiations were carried out on 3D spheroids maintained under a range of O2 and temperature conditions to control O2 consumption and create gradients representative of in vivo tissues. RESULTS: Staining for pDNA-PK (Ser2056) produced a linear radiation dose response whereas γH2AX (Ser139) showed saturation with increasing dose. Using the pDNA-PK staining, radiation response was then characterised for FLASH compared to standard-dose-rates as a function of depth into the spheroids. At 4 °C, chosen to minimize the development of metabolic oxygen gradients within the tissues, FLASH protection could be observed at all distances under oxygen conditions of 0.3-1 % O2. Whereas at 37 °C a FLASH-protective effect was limited to the outer cell layers of tissues, an effect only observed at 3 % O2. Modelling of changes in the pDNA-PK-based oxygen enhancement ratio (OER) yielded a tissue ROC g0-value estimate of 0.73 ± 0.25 µM/Gy with a km of 5.4 µM at FLASH dose rates. CONCLUSIONS: DNA damage response markers are sensitive to the effects of transient oxygen depletion during FLASH radiotherapy. Findings support the rationale that well-oxygenated tissues would benefit more from FLASH-dose-rate protection relative to poorly-oxygenated tissues.


Asunto(s)
Daño del ADN , Esferoides Celulares , Daño del ADN/efectos de la radiación , Humanos , Esferoides Celulares/efectos de la radiación , Histonas/metabolismo , Histonas/análisis , Consumo de Oxígeno/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Tratamientos Conservadores del Órgano/métodos
6.
Sci Rep ; 13(1): 12429, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37528151

RESUMEN

Type II topoisomerase (Top2) poisoning therapy is used to treat a broad range of cancers via induction of double strand breaks (DSBs) in cells undergoing replication and transcription. Preventing the repair of DSBs via inhibition of DNA-PK, an inhibitor of non-homologous end-joining (NHEJ), increases cell kill with Top2 poisons and has led to the initiation of several clinical trials. To elucidate the cellular mechanisms leading to synergistic activity of dual DNA-PK/Top2 inhibition we looked at their effects in cycling versus non-cycling cells, in 3D spheroids and in xenograft models. Combined DNA-PK/Top2 inhibition was found to not only increase the cell kill in proliferating cells, the cell population that is typically most vulnerable to Top2 poisoning, but also in non-proliferative but transcriptionally active cells. This effect was observed in both cancer and normal tissue models, killing more cells than high concentrations of etoposide alone. The combination treatment delayed tumor growth in mice compared to Top2 poisoning alone, but also led to increased toxicity. These findings demonstrate sensitization of Top2ß-expressing, non-cycling cells to Top2 poisoning by DNA-PK inhibition. Expansion of the target cell population of Top2 poison treatment to include non-proliferating cells via combination with DNA damage repair inhibitors has implications for efficacy and toxicity of these combinations, including for inhibitors of DNA-PK currently in clinical trial.


Asunto(s)
Proteínas de Unión al ADN , Neoplasias , Humanos , Animales , Ratones , Proteínas de Unión al ADN/genética , ADN-Topoisomerasas de Tipo II/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , Etopósido/farmacología , Isomerasas/genética , Proteína Quinasa Activada por ADN/genética , Neoplasias/tratamiento farmacológico , ADN , Inhibidores de Topoisomerasa II/farmacología
7.
Haematologica ; 95(12): 2157-60, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20713461

RESUMEN

Follicular lymphoma patients display heterogeneous overall survival and variable risk of transformation. Recent studies have highlighted the role of the microenvironment. The contribution of microvessel density to follicular lymphoma survival remains controversial. We used a quantitative tumor mapping approach to determine whether the degree of vascularization correlated with outcome in a uniformly treated cohort. Whole-tissue sections of diagnostic biopsies from 84 cases were stained for CD34 and tumor-to-vessel-distance that encompassed 90% of the tumor (TVD(90)) was determined using image analysis. Twenty-one cases with lower TVD(90) showed inferior overall survival (P=0.0001) and high risk of transformation (P=0.01). These cases significantly correlated with increased Lymphoma-Associated Macrophages (χ(2)=0.025). In multivariate analysis macrophages content, IPI and TVD(90) were independent predictors of overall survival (P=0.05, P=0.001 and P=0.01, respectively) and IPI and TVD(90) predicted risk of transformation (P=0.008 and P=0.08, respectively). Increased angiogenesis is an independent marker of inferior survival and may promote transformation.


Asunto(s)
Linfoma Folicular/patología , Microvasos/patología , Neovascularización Patológica , Adolescente , Adulto , Antígenos CD34/metabolismo , Transformación Celular Neoplásica , Supervivencia sin Enfermedad , Humanos , Inmunohistoquímica , Linfoma Folicular/terapia , Macrófagos/patología , Microvasos/metabolismo , Persona de Mediana Edad , Análisis Multivariante , Pronóstico , Factores de Riesgo , Resultado del Tratamiento , Adulto Joven
8.
Clin Cancer Res ; 14(7): 2171-9, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18381959

RESUMEN

PURPOSE: The high molecular weight and binding affinity of trastuzumab, a monoclonal antibody in use for treatment of breast cancers overexpressing human epidermal growth factor receptor type 2 (HER2), in combination with microenvironmental factors, may limit its distribution and efficacy. We assessed and mapped the distribution of systemically given, unlabeled trastuzumab at micrometer resolution in tumor xenografts using immunohistochemistry. EXPERIMENTAL DESIGN: Mice bearing MDA-435/LCC6(HER2) xenografts were given single doses of 4 or 20 mg/kg unlabeled trastuzumab with tumor harvest at various time points thereafter; bound trastuzumab was imaged directly in tumor cryosections using fluorescently tagged antihuman secondary antibodies. Combinations of additional markers, including HER2, 5-bromo-2-deoxyuridine, CD31, DioC(7)(3), desmin, and collagen IV were also mapped on the same tumor sections. RESULTS: Distribution of trastuzumab in MDA-435/LCC6(HER2) tumors is found to be heterogeneous, with tumor margins saturating more thoroughly in doses and times analyzed. Considerable intervessel heterogeneity is also seen. For example, in unsaturated tissues, there remain perfused vessels without any trastuzumab in addition to vessels with a few layers of positively stained perivascular cells, in addition to vessels with bound drug up to 150 microm away. This heterogeneity is independent of HER2 expression, microvessel density, and perfusion. A slightly greater proportion of vessels were associated with pericytes in sections with greater trastuzumab saturation, but this would not adequately account for observed heterogeneous trastuzumab distribution. CONCLUSIONS: Complete penetration of trastuzumab in tumor tissue was not seen in our study, leaving the possibility that inadequate distribution may represent a mechanism for resistance to trastuzumab.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Antineoplásicos/farmacocinética , Neoplasias de la Mama/tratamiento farmacológico , Procesamiento de Imagen Asistido por Computador , Receptor ErbB-2/metabolismo , Animales , Anticuerpos Monoclonales Humanizados , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Inmunohistoquímica , Ratones , Distribución Tisular , Trastuzumab , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Mol Cancer Ther ; 7(4): 749-58, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18375822

RESUMEN

Molecular mechanisms responsible for lymphoma resistance to apoptosis often involve the bcl-2 pathway. In this study, we investigated the cell signaling pathways activated in bcl-2-overexpressing human mantle cell lymphoma cell lines (JVM-2 and Z-138) that have been treated with oblimersen, a molecular gene silencing strategy that effectively suppresses bcl-2 in vitro and in vivo. Z-138 cells expressed higher levels of bcl-2 and were more sensitive to the effects of bcl-2 silencing, mediated by oblimersen or bcl-2 small interfering RNA, in vitro. Tumors derived following injection of Z-138 cells were sensitive to oblimersen as judged by decreases in tumor growth rate and decreases in cell proliferation (as measured by Ki-67). Immunohistochemistry and Western blot analysis of oblimersen-treated Z-138 tumors revealed a dose-dependent decrease in bcl-2 levels and an associated increase in the proapoptotic proteins caspase-3 and caspase-9. Silencing bcl-2 in Z-138 xenografts revealed an associated dose-dependent suppression of bax, a decrease in nuclear factor-kappaB and phospho-nuclear factor-kappaB, and transient loss of p53 levels. Coimmunoprecipitation studies suggest that the latter observation is mediated by an association between bcl-2 and phospho-mdm2. Bcl-2 silencing also led to p27 down-regulation and coimmunoprecipitation studies point to a role for bcl-2 in regulation of p27 localization/degradation. Bcl-2 silencing was also correlated with loss of cyclin D1a protein levels but not cyclin D1b levels. Coimmunoprecipitation studies indicate that bcl-2 may mediate its effects on cyclin D1a via interaction with p38 mitogen-activated protein kinase as well as a previously unreported interaction between bcl-2 and cyclin D1a.


Asunto(s)
Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Silenciador del Gen , Linfoma de Células del Manto/genética , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Animales , Apoptosis/fisiología , Western Blotting , Proliferación Celular , Ciclina D , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Proteínas de Unión al ADN/fisiología , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Linfoma de Células del Manto/metabolismo , Linfoma de Células del Manto/prevención & control , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas de Neoplasias/genética , Oligonucleótidos Antisentido/farmacología , Fosforilación , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tionucleótidos/farmacología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
10.
Mol Cancer Ther ; 7(1): 59-70, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18202010

RESUMEN

Integrin-linked kinase (ILK) was assesed as a therapeutic target in glioblastoma xenograft models through multiple endpoints including treatment related changes in the tumor microenvironment. Glioblastoma cell lines were tested in vitro for sensitivity toward the small-molecule inhibitors QLT0254 and QLT0267. Cell viability, cell cycle, and apoptosis were evaluated using MTT assay, flow cytometry, caspase activation, and DAPI staining. Western blotting and ELISA were used for protein analysis (ILK, PKB/Akt, VEGF, and HIF-1alpha). In vivo assessment of growth rate, cell proliferation, BrdUrd, blood vessel mass (CD31 labeling), vessel perfusion (Hoechst 33342), and hypoxia (EF-5) was done using U87MG glioblastoma xenografts in RAG2-M mice treated orally with QLT0267 (200 mg/kg q.d.). ILK inhibition in vitro with QLT0254 and QLT0267 resulted in decreased levels of phospho-PKB/Akt (Ser473), secreted VEGF, G2-M block, and apoptosis induction. Mice treated with QLT0267 exhibited significant delays in tumor growth (treated 213 mm3 versus control 549 mm3). In situ analysis of U87MG tumor cell proliferation from QLT0267-treated mice was significantly lower relative to untreated mice. Importantly, VEGF and HIF-1alpha expression decreased in QLT0267-treated tumors as did the percentage of blood vessel mass and numbers of Hoechst 33342 perfused tumor vessels compared with control tumors (35% versus 83%). ILK inhibition with novel small-molecule inhibitors leads to treatment-associated delays in tumor growth, decreased tumor angiogenesis, and functionality of tumor vasculature. The therapeutic effects of a selected ILK inhibitor (QLT0267) should be determined in the clinic in cancers that exhibit dysregulated ILK, such as PTEN-null glioblastomas.


Asunto(s)
Glioblastoma/enzimología , Glioblastoma/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Glioblastoma/irrigación sanguínea , Glioblastoma/tratamiento farmacológico , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Estructura Molecular , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Immunol Res ; 7(7): 1064-1078, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31088846

RESUMEN

Treatment strategies involving immune-checkpoint blockade (ICB) have significantly improved survival for a subset of patients across a broad spectrum of advanced solid cancers. Despite this, considerable room for improving response rates remains. The tumor microenvironment (TME) is a hurdle to immune function, as the altered metabolism-related acidic microenvironment of solid tumors decreases immune activity. Here, we determined that expression of the hypoxia-induced, cell-surface pH regulatory enzyme carbonic anhydrase IX (CAIX) is associated with worse overall survival in a cohort of 449 patients with melanoma. We found that targeting CAIX with the small-molecule SLC-0111 reduced glycolytic metabolism of tumor cells and extracellular acidification, resulting in increased immune cell killing. SLC-0111 treatment in combination with immune-checkpoint inhibitors led to the sensitization of tumors to ICB, which led to an enhanced Th1 response, decreased tumor growth, and reduced metastasis. We identified that increased expression of CA9 is associated with a reduced Th1 response in metastatic melanoma and basal-like breast cancer TCGA cohorts. These data suggest that targeting CAIX in the TME in combination with ICB is a potential therapeutic strategy for enhancing response and survival in patients with hypoxic solid malignancies.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Anhidrasas Carbónicas/química , Hipoxia/fisiopatología , Neoplasias Pulmonares/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Compuestos de Fenilurea/farmacología , Sulfonamidas/farmacología , Animales , Apoptosis , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Antígeno CTLA-4/antagonistas & inhibidores , Anhidrasas Carbónicas/metabolismo , Proliferación Celular , Quimioterapia Combinada , Inducción Enzimática , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/secundario , Melanoma/enzimología , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Pronóstico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Tasa de Supervivencia , Células Tumorales Cultivadas , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
12.
Clin Cancer Res ; 13(9): 2804-10, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17473214

RESUMEN

PURPOSE: Limited drug penetration in solid tumors is a potential mechanism of resistance for many anticancer drugs. Taxanes represent a class of drugs that are currently undergoing a new round of development, but with little known of their ability to penetrate and distribute relative to blood vessels within solid tumors. EXPERIMENTAL DESIGN: We assessed the tissue penetration of paclitaxel and docetaxel in HCT-116 tumor xenografts and in multilayered cell culture (MCC), a three-dimensional cell culture model of the tumor extravascular compartment. In xenografts, taxanes were mapped relative to blood vessels to obtain drug profiles as a function of distance from vasculature. For MCC, cultures were exposed to stirred drug reservoirs and taxanes measured as a function of depth into tissue. RESULTS: Both taxanes exhibited limited penetration, with little drug reaching further than 100 microm into the tissue. Of the two, paclitaxel exhibited up to 2-fold greater penetration than docetaxel. Mapping tumor cell proliferation following treatment allowed the consequences of limited drug penetration to be assessed. In tumor xenografts where reduced drug exposure to cells far from vasculature is one of several factors influencing response to treatment, up to a 75% reduction in S-phase cells was achieved in cells nearest the vessels, but only 50% reduction was observed in the tissue 150 microm away. In MCC-based data, where the influence of reduced cell proliferation with depth into tissue was circumvented, a 5-fold (paclitaxel) and 10-fold (docetaxel) increase in reservoir drug concentration was required to produce a response in cells 150 microm into the tissue equivalent to that seen in cells directly exposed to the drug. CONCLUSION: These results indicate that limited distribution is an important mechanism of tumor resistance to taxanes.


Asunto(s)
Antineoplásicos/farmacocinética , Resistencia a Antineoplásicos , Neoplasias/metabolismo , Paclitaxel/farmacología , Taxoides/farmacocinética , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Docetaxel , Humanos , Ratones , Ratones Endogámicos , Fase S/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Radiother Oncol ; 78(2): 138-45, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16455148

RESUMEN

BACKGROUND AND PURPOSE: Tirapazamine is a hypoxic cytotoxin currently undergoing Phase II/III clinical evaluation in combination with radiation and chemotherapeutics for the treatment of non-hematological cancers. Tissue penetration studies using multicellular models have suggested that tirapazamine exposure may be limited to cells close to blood vessels. However, animal studies show tirapazamine enhances the anti-tumour activity of radiation and chemotherapy and clinical studies with tirapazamine, so far, are promising. To investigate this apparent paradox we examined the microregional effects of tirapazamine in vivo by mapping drug effects with respect to the position of blood vessels in tumour cryosections. PATIENTS AND METHODS: Tirapazamine was administered i.p. to mice bearing HCT-116 tumours, which were excised at various times after treatment. Images of multiple-stained cryosections were overlaid to provide microregional information on the relative position of proliferating cells, hypoxia, perfusion and vasculature. RESULTS: We observed extensive and permanent vascular dysfunction in a large proportion of tumours from mice treated with tirapazamine. In the affected tumours, blood flow ceased in the centrally located tumour vessels, leaving a rim of functional vessels around the periphery of the tumour. This vascular dysfunction commenced within 24 h after tirapazamine administration and the areas affected appeared to be replaced by necrosis over the following 24-48 h. CONCLUSIONS: Because the majority of hypoxic cells are located in the center of tumours we propose that the activity of tirapazamine in vivo may be related to its effects on tumour vasculature and that its activity against hypoxic cells located distal to functional blood vessels may not be as important as previously believed.


Asunto(s)
Antineoplásicos/uso terapéutico , Células HCT116/trasplante , Neoplasias Cutáneas/irrigación sanguínea , Trasplante Heterólogo , Triazinas/uso terapéutico , Animales , Antimetabolitos , Vasos Sanguíneos/efectos de los fármacos , Bromodesoxiuridina , Hipoxia de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colorantes , Femenino , Células HCT116/efectos de los fármacos , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Ratones , Ratones Endogámicos NOD , Ratones SCID , Necrosis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta , Flujo Sanguíneo Regional/efectos de los fármacos , Neoplasias Cutáneas/tratamiento farmacológico , Tirapazamina
14.
Cancer Res ; 64(18): 6537-41, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15374965

RESUMEN

To examine the tumor microregional effects after gemcitabine administration to mice, we mapped the location of proliferating and hypoxic cells relative to vasculature in human colon cancer xenografts. The S-phase marker bromodeoxyuridine was used as a surrogate of drug effect and administered 2 hours before tumor excision, whereas vessel position and perfusion were assessed via staining for CD31 and intravenous injection of carbocyanine, respectively. Hypoxia was detected using pimonidazole. Images of the four markers were overlaid to reveal the spatial relationship between proliferation, vasculature, and hypoxia and to examine the microregional effects. Within 1 day after administration of 240 mg/kg of gemcitabine, proliferation throughout the tumor was completely inhibited. Over time, a reemergence of dividing cells occurred in relation to the distance from vasculature. Microregional analysis revealed that cells located distal to vasculature commenced cycling sooner than cells located proximal to vasculature. A similar trend was seen after multiple doses of gemcitabine (40 mg/kg on days 1, 4, 7, and 10). The possibility that the effect of gemcitabine could be attributed to changes in oxygenation was discounted after examining the vessel perfusion and patterns of hypoxia. The effect of gemcitabine was examined in multilayered cell culture, and at doses <30 micromol/L, a gradient in proliferation between the exposed and unexposed sides was observed. We show a differential effect on cell proliferation in relation to vasculature and conclude that cells distal to blood vessels are less affected by gemcitabine probably because of limited penetration.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Animales , Antimetabolitos Antineoplásicos/farmacocinética , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/metabolismo , Desoxicitidina/farmacocinética , Femenino , Células HCT116 , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
15.
Cancer Res ; 64(17): 6304-9, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15342419

RESUMEN

The failure of many anticancer drugs to control growth of solid cancers may stem in part from inadequate delivery to tumor regions distant from vasculature. Although the identification of new anticancer drug targets has led to the development of many new drug candidates, there is a lack of methodology for identifying drugs that adequately penetrate tumor tissue. We have developed a novel multilayered cell culture-based assay, which detects the penetration of anticancer drugs based on their effect within tissue. Drug exposures are made over 1 hour to one side of a disk of tissue approximately 150-microm thick, with the other side temporarily closed off, and penetration is then assessed 1-3 days later via bromodeoxyuridine-based detection of S-phase cells. Using this assay, the tissue distribution of a selection of anthracycline analogues was assessed. At clinically relevant exposures, none of the agents were able to affect cells on the far side of the culture at levels approaching that seen on the near (exposed) side. Doxorubicin and epirubicin exhibited approximately 10-fold decreases in the drug exposure seen by the cells on the far side relative to those on the near side of the cultures, whereas for daunorubicin and mitoxantrone, approximately 30-fold and >30-fold decreases were observed respectively. Results were consistent with the observed gradients in drug-derived fluorescence of doxorubicin, epirubicin, and daunorubicin. This model could be applied as a simple anticancer drug development screen to discover drugs that exhibit desirable penetration properties.


Asunto(s)
Antraciclinas/farmacocinética , Antibióticos Antineoplásicos/farmacocinética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Daunorrubicina/farmacocinética , Doxorrubicina/farmacocinética , Epirrubicina/farmacocinética , Células HCT116 , Humanos , Concentración de Iones de Hidrógeno , Cinética , Mitoxantrona/farmacocinética , Espectrometría de Fluorescencia , Células Tumorales Cultivadas
16.
Cancer Res ; 63(18): 5707-11, 2003 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-14522888

RESUMEN

Bromodeoxyuridine (BrdUrd) is used extensively to measure the fraction of proliferating cells in tumors. Unlike endogenous markers of proliferation such as proliferating cell nuclear antigen (PCNA) and Ki-67, BrdUrd is exogenously administered and reaches the tumor via vasculature where it must then diffuse throughout the tissue to label S-phase cells. In this study, we examine the dose dependence of BrdUrd on the tumor distribution of labeled cells in histological sections. Analysis of the distribution of labeled cells in SiHa tumor xenografts showed that a dose between 400 and 1000 mg/kg was required to label cells 150 micro m from blood vessels, approaching the border of necrosis. Lower doses resulted in only the cells close to blood vessels being labeled. Interestingly, cells residing furthest from blood vessels still labeled albeit at half the level of cells situated proximal to the tumor vasculature. Results were compared with the penetration of BrdUrd seen in vitro using multilayered cell culture (MCC), a three-dimensional tissue culture model of solid tumors. Using MCC, an exposure of 100 micro M BrdUrd for 1 h was required for labeling of S-phase cells 150 micro m into the tissue, whereas cells adjacent to the edge of the tissue could be adequately labeled with just 5 micro M BrdUrd for 1 h. The area under the curve for a 100 mg/kg BrdUrd dose in mice was found to be approximately 30 micro M x h.


Asunto(s)
Bromodesoxiuridina/farmacocinética , Carcinoma de Células Escamosas/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Animales , Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/patología , División Celular/fisiología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Trasplante Heterólogo , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/irrigación sanguínea , Neoplasias del Cuello Uterino/patología
17.
Mol Cancer Res ; 2(11): 606-19, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15561777

RESUMEN

The effects of HER-2/neu overexpression on the tumor microenvironment in an aggressive breast cancer xenograft model were investigated. These studies focused on tumors derived following the subcutaneous injection of MDA-MB-435/LCC6 cells transfected with human c-erbB2 (LCC6(HER-2)) into SCID-Rag2M mice. LCC6(HER-2) tumors were more viable (H&E-stained tumor sections) than isogenic vector control tumors (LCC6(Vector)). Correspondingly, a 2.7-fold increase in trypan blue-excluding cells (P = 0.00056) and a 4.8-fold increase in clonogenic cells (P = 0.00146) were noted in cell suspensions derived from disaggregated LCC6(HER-2) versus LCC6(Vector) tumors. Tumor sections stained with the antibody detecting 2-(2-nitro-1H-imidazol-1-yl)-N-(2,2,3,3,3-pentafluoropropyl)-acetamide (EF5), a marker of hypoxia, showed a greater fraction of hypoxic tissue in LCC6(HER-2) tumors compared with control tumors. Flow cytometric analyses based on viable tumor cells (DNA content >/= 2N) in cell suspensions from disaggregated tumors confirmed that there were significantly more EF5-positive cells (i.e., hypoxic) in LCC6(HER-2) than in LCC6(Vector) tumors (16.41 +/- 8.1% and 5.96 +/- 4.1%, respectively; P = 0.0015). Protein levels of phosphorylated (Ser(536)) nuclear factor-kappaB p65 were significantly elevated in LCC6(HER-2) tumors (P = 0.00048), and a trend in increased hypoxia-inducible factor-1alpha protein levels was observed in LCC6(HER-2) compared with LCC6(Vector) tumors. Despite the substantial viable hypoxic cell fraction and a 1.7-fold increase of vascular endothelial growth factor protein (P = 0.05) in LCC6(HER-2) tumors, no significant differences were found (P > 0.05) between LCC6(HER-2) and LCC6(Vector) vasculature (CD31 staining and Hoechst 33342 perfusion). These results suggest that HER-2/neu overexpression may be linked with overall increased tumor viability and a significant increase in the population of viable hypoxic cells, which is not due to differences in tumor vascularization.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Etanidazol/análogos & derivados , Neovascularización Patológica/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Adaptación Fisiológica/genética , Animales , Biomarcadores , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/genética , Carcinoma/irrigación sanguínea , Carcinoma/genética , Hipoxia de la Célula/genética , Línea Celular Tumoral/trasplante , Proliferación Celular , Respiración de la Célula/genética , Supervivencia Celular/genética , Células Clonales/metabolismo , Resistencia a Antineoplásicos/genética , Femenino , Supervivencia de Injerto/fisiología , Humanos , Hidrocarburos Fluorados , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ratones , Ratones SCID , FN-kappa B/metabolismo , Factor de Transcripción ReIA , Factores de Transcripción/metabolismo , Trasplante Heterólogo , Regulación hacia Arriba/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Cancer Cell ; 27(5): 682-97, 2015 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-25965573

RESUMEN

Metastatic dissemination is the leading cause of death in cancer patients, which is particularly evident for high-risk sarcomas such as Ewing sarcoma, osteosarcoma, and rhabdomyosarcoma. Previous research identified a crucial role for YB-1 in the epithelial-to-mesenchymal transition (EMT) and metastasis of epithelial malignancies. Based on clinical data and two distinct animal models, we now report that YB-1 is also a major metastatic driver in high-risk sarcomas. Our data establish YB-1 as a critical regulator of hypoxia-inducible factor 1α (HIF1α) expression in sarcoma cells. YB-1 enhances HIF1α protein expression by directly binding to and activating translation of HIF1A messages. This leads to HIF1α-mediated sarcoma cell invasion and enhanced metastatic capacity in vivo, highlighting a translationally regulated YB-1-HIF1α axis in sarcoma metastasis.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Metástasis de la Neoplasia , Biosíntesis de Proteínas , Sarcoma/patología , Proteína 1 de Unión a la Caja Y/fisiología , Humanos , Invasividad Neoplásica , Sarcoma/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/fisiología
19.
Clin Cancer Res ; 21(15): 3428-35, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25869385

RESUMEN

PURPOSE: We aimed to assess the prognostic significance of follicular lymphoma-associated macrophages in the era of rituximab treatment and maintenance. EXPERIMENTAL DESIGN: We applied immunohistochemistry for CD68 and CD163 to two large tissue microarrays (TMA). The first TMA included samples from 186 patients from the BC Cancer Agency (BCCA) who had been treated with first-line systemic treatment including rituximab, cyclophosphamide, vincristine, and prednisone. The second contained 395 samples from PRIMA trial patients treated with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone, and randomized to rituximab maintenance or observation. Macrophage infiltration was assessed using Aperio image analysis. Each of the two cohorts was randomly split into training/validation sets. RESULTS: An increased CD163-positive pixel count was predictive of adverse outcome in the BCCA dataset [5-year progression-free survival (PFS) 38% vs. 72%, respectively, P = 0.004 in the training cohort and 5-year PFS 29% vs. 61%, respectively, P = 0.004 in the validation cohort]. In the PRIMA trial, an increased CD163 pixel count was associated with favorable outcome (5-year PFS 60% vs. 44%, respectively, P = 0.011 in the training cohort and 5-year PFS 55% vs. 37%, respectively, P = 0.030 in the validation cohort). CONCLUSIONS: CD163-positive macrophages predict outcome in follicular lymphoma, but their prognostic impact is highly dependent on treatment received.


Asunto(s)
Antígenos CD/biosíntesis , Antígenos de Diferenciación Mielomonocítica/biosíntesis , Linfoma Folicular/tratamiento farmacológico , Linfoma Folicular/genética , Pronóstico , Receptores de Superficie Celular/biosíntesis , Rituximab/administración & dosificación , Anciano , Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/genética , Ciclofosfamida/administración & dosificación , Supervivencia sin Enfermedad , Doxorrubicina/administración & dosificación , Femenino , Humanos , Linfoma Folicular/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Receptores de Superficie Celular/genética , Análisis de Matrices Tisulares , Resultado del Tratamiento , Microambiente Tumoral/efectos de los fármacos , Vincristina/administración & dosificación
20.
Radiat Res ; 158(1): 94-100, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12071808

RESUMEN

The penetration of anticancer agents into tumor tissue has recently attracted considerable attention. This study examines the effect of carbogen breathing on the antitumor activity of tirapazamine combined with radiation. Our hypothesis is based on the observation that the diffusion of tirapazamine through tissue is dependent on oxygen tension. We postulated that carbogen breathing might enhance the ability of tirapazamine to diffuse to hypoxic cells located distal to functional blood vessels in tumors. We first determined that carbogen breathing caused no significant change in the pharmacokinetics of tirapazamine, suggesting that any effect of carbogen breathing on the activity of tirapazamine is not attributable to modulation of pharmacokinetics. Cell survival in SCCVII and SiHa tumors after 10 Gy X rays alone was similar. However, when tirapazamine was administered 30 min after radiation treatment under air-breathing conditions, cell killing was greater in SCCVII tumors compared to SiHa tumors. Carbogen breathing during the exposure to tirapazamine did not change the cell survival in SCCVII tumors, but it enhanced cell killing in the SiHa tumors. Interestingly, carbogen breathing during radiation treatment produced greater cell killing in the SiHa tumors than in the SCCVII tumors. The vascular architecture and type of hypoxia in the two tumors probably underlie the differences in the responses of the two tumors. These findings suggest that the effectiveness of tirapazamine and other hypoxic cytotoxins may be dependent on tumor type.


Asunto(s)
Dióxido de Carbono/toxicidad , Carcinoma de Células Escamosas/terapia , Rayos gamma , Oxígeno/toxicidad , Fármacos Sensibilizantes a Radiaciones/toxicidad , Triazinas/toxicidad , Neoplasias del Cuello Uterino/terapia , Administración por Inhalación , Animales , Dióxido de Carbono/administración & dosificación , Carcinoma de Células Escamosas/patología , Sinergismo Farmacológico , Femenino , Humanos , Ratones , Ratones Endogámicos C3H , Ratones SCID , Oxígeno/administración & dosificación , Tirapazamina , Neoplasias del Cuello Uterino/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA