Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Dig Dis Sci ; 68(7): 2975-2980, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36884186

RESUMEN

BACKGROUND: Pentraxin-2 (PTX-2) is a homo-pentameric plasma protein showing evidence of antifibrotic activity in Phase 2 clinical trials in idiopathic pulmonary fibrosis (IPF). Whether PTX-2 plays a role in other fibrotic diseases, including intestinal fibrosis which commonly occurs in inflammatory bowel disease (IBD), remains unknown. AIMS: This study aimed to qualitatively and quantitatively assess PTX-2 expression in fibrostenotic Crohn's disease (FCD) and determine whether expression is correlated with postsurgical restenosis. METHODS: Immunohistochemistry was performed in histologic sections of small bowel resected from patients with fibrostenotic Crohn's disease (FCD), comparing strictured segments with adjacent surgical margins from the same patient. Ileal resections from patients without inflammatory bowel disease were examined as controls. RESULTS: PTX-2 signal was analyzed in 18 patients with FCD and 15 patients without IBD and localized predominantly to submucosal vasculature, including arterial subendothelium and internal elastic lamina, and perivascular connective tissue. PTX-2 signal in the surgical margins from patients with FCD strictures (where tissue architecture was normal) was consistently lower than non-IBD samples. Fibrostenotic regions showed increased PTX-2 signal relative to surgical margins from the same patient in 14/15 paired samples. Submucosal/mural PTX-2 signal in fibrostenotic tissue was lower in patients who subsequently experienced re-stenosis (P = 0.015). CONCLUSIONS: This exploratory study is the first analysis of PTX-2 within the intestine, and demonstrates that PTX-2 signal is reduced in the architecturally normal bowel of patients with FCD. Lower submucosal PTX-2 levels in patients with re-stenosis raises the possibility of a protective role of PTX-2 in intestinal fibrosis.


Asunto(s)
Enfermedad de Crohn , Enfermedades Inflamatorias del Intestino , Humanos , Constricción Patológica/patología , Enfermedad de Crohn/complicaciones , Enfermedad de Crohn/cirugía , Enfermedad de Crohn/metabolismo , Fibrosis , Enfermedades Inflamatorias del Intestino/patología , Intestinos/patología , Márgenes de Escisión
2.
J Vis Exp ; (174)2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34487120

RESUMEN

Immunohistochemistry (IHC) assays provide valuable insights into protein expression patterns, the reliable interpretation of which requires well-characterized positive and negative control samples. Because appropriate tissue or cell line controls are not always available, a simple method to create synthetic IHC controls may be beneficial. Such a method is described here. It is adaptable to various antigen types, including proteins, peptides, or oligonucleotides, in a wide range of concentrations. This protocol explains the steps necessary to create synthetic antigen controls, using as an example a peptide from the human erythroblastic oncogene B2 (ERBB2/HER2) intracellular domain (ICD) recognized by a variety of diagnostically relevant antibodies. Serial dilutions of the HER2 ICD peptide in bovine serum albumin (BSA) solution are mixed with formaldehyde and heated for 10 min at 85 °C to solidify and cross-link the peptide/BSA mixture. The resulting gel can be processed, sectioned, and stained like a tissue, yielding a series of samples of known antigen concentrations spanning a wide range of staining intensities. This simple protocol is consistent with routine histology lab procedures. The method requires only that the user have a sufficient quantity of the desired antigen. Recombinant proteins, protein domains, or linear peptides that encode relevant epitopes may be synthesized locally or commercially. Laboratories generating in-house antibodies can reserve aliquots of the immunizing antigen as the synthetic control target. The opportunity to create well-defined positive controls across a wide range of concentrations allows users to assess intra- and inter-laboratory assay performance, gain insight into the dynamic range and linearity of their assays, and optimize assay conditions for their particular experimental goals.


Asunto(s)
Antígenos , Formaldehído , Epítopos , Humanos , Inmunohistoquímica , Vacunas Sintéticas
4.
Clin Lymphoma Myeloma Leuk ; 21(4): 267-278.e10, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33303421

RESUMEN

INTRODUCTION: The prognostic value of B-cell lymphoma 2 (BCL2) expression in de novo diffuse large B-cell lymphoma (DLBCL) treated with immunochemotherapy is of interest to define a target patient population for clinical development of BCL2 inhibitors. We aimed to develop a reproducible immunohistochemistry algorithm and assay to determine BCL2 protein expression and assess the prognostic value of BCL2 in newly diagnosed DLBCL cohorts. PATIENTS AND METHODS: The prospectively defined algorithm incorporated BCL2 staining intensity and percentage of BCL2-positive cells. Functionally relevant cutoffs were based on the sensitivity of lymphoma cell lines to venetoclax. This assay was highly reproducible across laboratories. The prognostic impact of BCL2 expression was assessed in DLBCL patients from the phase 3 MAIN (n = 230) and GOYA (n = 366) trials, and a population-based registry (n = 310). RESULTS: Approximately 50% of tumors were BCL2 positive, with a higher frequency in high International Prognostic Index (IPI) and activated B-cell-like DLBCL subgroups. BCL2 expression was associated with poorer progression-free survival in the MAIN study (hazard ratio [HR], 1.66; 95% confidence interval [CI], 0.81-3.40; multivariate Cox regression adjusted for IPI and cell of origin). This trend was confirmed in the GOYA and registry cohorts in adjusted multivariate analyses (GOYA: HR, 1.72; 95% CI, 1.05-2.82; registry: HR, 1.89; 95% CI, 1.29-2.78). Patients with BCL2 immunohistochemistry-positive and IPI-high disease had the poorest prognosis: 3-year progression-free survival rates were 51% (GOYA) and 37% (registry). CONCLUSION: Findings support use of our BCL2 immunohistochemistry scoring system and assay to select patients with BCL2-positive tumors for future studies.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/genética , Linfoma de Células B Grandes Difuso/mortalidad , Proteínas Proto-Oncogénicas c-bcl-2/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Biomarcadores de Tumor/análisis , Ensayos Clínicos Fase III como Asunto , Estudios de Cohortes , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Estimación de Kaplan-Meier , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Masculino , Persona de Mediana Edad , Estudios Multicéntricos como Asunto , Pronóstico , Supervivencia sin Progresión , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Ensayos Clínicos Controlados Aleatorios como Asunto , Sistema de Registros/estadística & datos numéricos , Adulto Joven
5.
J Histochem Cytochem ; 67(5): 309-334, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30879407

RESUMEN

Optimization and standardization of immunohistochemistry (IHC) protocols within and between laboratories requires reproducible positive and negative control samples. In many situations, suitable tissue or cell line controls are not available. We demonstrate here a method to incorporate target antigens into synthetic protein gels that can serve as IHC controls. The method can use peptides, protein domains, or whole proteins as antigens, and is compatible with a variety of fixation protocols. The resulting gels can be used to create tissue microarrays (TMAs) with a range of antigen concentrations that can be used to objectively quantify and calibrate chromogenic, fluorescent, or mass spectrometry-based IHC protocols. The method offers an opportunity to objectively quantify IHC staining results, and to optimize and standardize IHC protocols within and between laboratories. (J Histochem Cytochem 58:XXX-XXX, 2019).


Asunto(s)
Antígenos/análisis , Geles/química , Inmunohistoquímica/métodos , Animales , Formaldehído/química , Humanos , Inmunohistoquímica/normas , Ratones , Coloración y Etiquetado/métodos , Coloración y Etiquetado/normas , Análisis de Matrices Tisulares/métodos , Análisis de Matrices Tisulares/normas , Fijación del Tejido/métodos , Fijación del Tejido/normas
6.
Neoplasia ; 19(11): 950-959, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28987998

RESUMEN

The effect of anti-angiogenic agents on tumor oxygenation has been in question for a number of years, where both increases and decreases in tumor pO2 have been observed. This dichotomy in results may be explained by the role of vessel normalization in the response of tumors to anti-angiogenic therapy, where anti-angiogenic therapies may initially improve both the structure and the function of tumor vessels, but more sustained or potent anti-angiogenic treatments will produce an anti-vascular response, producing a more hypoxic environment. The first goal of this study was to employ multispectral (MS) 19F-MRI to noninvasively quantify viable tumor pO2 and evaluate the ability of a high dose of an antibody to vascular endothelial growth factor (VEGF) to produce a strong and prolonged anti-vascular response that results in significant tumor hypoxia. The second goal of this study was to target the anti-VEGF induced hypoxic tumor micro-environment with an agent, tirapazamine (TPZ), which has been designed to target hypoxic regions of tumors. These goals have been successfully met, where an antibody that blocks both murine and human VEGF-A (B20.4.1.1) was found by MS 19F-MRI to produce a strong anti-vascular response and reduce viable tumor pO2 in an HM-7 xenograft model. TPZ was then employed to target the anti-VEGF-induced hypoxic region. The combination of anti-VEGF and TPZ strongly suppressed HM-7 tumor growth and was superior to control and both monotherapies. This study provides evidence that clinical trials combining anti-vascular agents with hypoxia-activated prodrugs should be considered to improved efficacy in cancer patients.


Asunto(s)
Imagen por Resonancia Magnética con Fluor-19/métodos , Hipoxia Tumoral/fisiología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Femenino , Células HT29 , Humanos , Ratones , Ratones Desnudos , Tirapazamina , Triazinas/farmacología , Triazinas/uso terapéutico , Hipoxia Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
7.
Clin Cancer Res ; 23(10): 2423-2432, 2017 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-27815358

RESUMEN

Purpose: Chk1 inhibition potentiates DNA-damaging chemotherapy by overriding cell-cycle arrest and genome repair. This phase I study evaluated the Chk1 inhibitor GDC-0425 given in combination with gemcitabine to patients with advanced solid tumors.Experimental Design: Patients received GDC-0425 alone for a 1-week lead-in followed by 21-day cycles of gemcitabine plus GDC-0425. Gemcitabine was initially administered at 750 mg/m2 (Arm A), then increased to 1,000 mg/m2 (Arm B), on days 1 and 8 in a 3 + 3 + 3 dose escalation to establish maximum tolerated dose (MTD). GDC-0425 was initially administered daily for three consecutive days; however, dosing was abbreviated to a single day on the basis of pharmacokinetics and tolerability. TP53 mutations were evaluated in archival tumor tissue. On-treatment tumor biopsies underwent pharmacodynamic biomarker analyses.Results: Forty patients were treated with GDC-0425. The MTD of GDC-0425 was 60 mg when administered approximately 24 hours after gemcitabine 1,000 mg/m2 Dose-limiting toxicities included thrombocytopenia (n = 5), neutropenia (n = 4), dyspnea, nausea, pyrexia, syncope, and increased alanine aminotransferase (n = 1 each). Common related adverse events were nausea (48%); anemia, neutropenia, vomiting (45% each); fatigue (43%); pyrexia (40%); and thrombocytopenia (35%). The GDC-0425 half-life was approximately 15 hours. There were two confirmed partial responses in patients with triple-negative breast cancer (TP53-mutated) and melanoma (n = 1 each) and one unconfirmed partial response in a patient with cancer of unknown primary origin.Conclusions: Chk1 inhibition with GDC-0425 in combination with gemcitabine was tolerated with manageable bone marrow suppression. The observed preliminary clinical activity warrants further investigation of this chemopotentiation strategy. Clin Cancer Res; 23(10); 2423-32. ©2016 AACR.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Melanoma/tratamiento farmacológico , Piperidinas/administración & dosificación , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Desoxicitidina/administración & dosificación , Desoxicitidina/efectos adversos , Desoxicitidina/análogos & derivados , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Femenino , Compuestos Heterocíclicos con 3 Anillos/efectos adversos , Humanos , Masculino , Melanoma/genética , Melanoma/patología , Persona de Mediana Edad , Piperidinas/efectos adversos , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Gemcitabina
8.
Neoplasia ; 15(11): 1241-50, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24339736

RESUMEN

Quantifying oxygenation in viable tumor remains a major obstacle toward a better understanding of the tumor micro-environment and improving treatment strategies. Current techniques are often complicated by tumor heterogeneity. Herein, a novel in vivo approach that combines (19)F magnetic resonance imaging ((19)F-MRI) R 1 mapping with diffusion-based multispectral (MS) analysis is introduced. This approach restricts the partial pressure of oxygen (pO2) measurements to viable tumor, the tissue of therapeutic interest. The technique exhibited sufficient sensitivity to detect a breathing gas challenge in a xenograft tumor model, and the hypoxic region measured by MS (19)F-MRI was strongly correlated with histologic estimates of hypoxia. This approach was then applied to address the effects of antivascular agents on tumor oxygenation, which is a research question that is still under debate. The technique was used to monitor longitudinal pO2 changes in response to an antibody to vascular endothelial growth factor (B20.4.1.1) and a selective dual phosphoinositide 3-kinase/mammalian target of rapamycin inhibitor (GDC-0980). GDC-0980 reduced viable tumor pO2 during a 3-day treatment period, and a significant reduction was also produced by B20.4.1.1. Overall, this method provides an unprecedented view of viable tumor pO2 and contributes to a greater understanding of the effects of antivascular therapies on the tumor's microenvironment.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Oxígeno/metabolismo , Animales , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Femenino , Xenoinjertos , Humanos , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Desnudos , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Microambiente Tumoral/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Nat Med ; 19(9): 1114-23, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23913124

RESUMEN

Although angiogenesis inhibitors have provided substantial clinical benefit as cancer therapeutics, their use is limited by resistance to their therapeutic effects. While ample evidence indicates that such resistance can be influenced by the tumor microenvironment, the underlying mechanisms remain incompletely understood. Here, we have uncovered a paracrine signaling network between the adaptive and innate immune systems that is associated with resistance in multiple tumor models: lymphoma, lung and colon. Tumor-infiltrating T helper type 17 (T(H)17) cells and interleukin-17 (IL-17) induced the expression of granulocyte colony-stimulating factor (G-CSF) through nuclear factor κB (NF-κB) and extracellular-related kinase (ERK) signaling, leading to immature myeloid-cell mobilization and recruitment into the tumor microenvironment. The occurrence of T(H)17 cells and Bv8-positive granulocytes was also observed in clinical tumor specimens. Tumors resistant to treatment with antibodies to VEGF were rendered sensitive in IL-17 receptor (IL-17R)-knockout hosts deficient in T(H)17 effector function. Furthermore, pharmacological blockade of T(H)17 cell function sensitized resistant tumors to therapy with antibodies to VEGF. These findings indicate that IL-17 promotes tumor resistance to VEGF inhibition, suggesting that immunomodulatory strategies could improve the efficacy of anti-angiogenic therapy.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Resistencia a Antineoplásicos , Interleucina-17/metabolismo , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/inmunología , Células Th17/inmunología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Anticuerpos/inmunología , Antígeno CD11b/metabolismo , Neoplasias Colorrectales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Fibroblastos/metabolismo , Hormonas Gastrointestinales/metabolismo , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Granulocitos/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Linfoma/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , FN-kappa B/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Neuropéptidos/metabolismo , Comunicación Paracrina , Microambiente Tumoral/inmunología , Factor A de Crecimiento Endotelial Vascular/inmunología
10.
Proc Natl Acad Sci U S A ; 110(15): 6079-84, 2013 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-23530240

RESUMEN

Granulocyte-colony stimulating factor (G-CSF) promotes mobilization of CD11b(+)Gr1(+) myeloid cells and has been implicated in resistance to anti-VEGF therapy in mouse models. High G-CSF production has been associated with a poor prognosis in cancer patients. Here we show that activation of the RAS/MEK/ERK pathway regulates G-CSF expression through the Ets transcription factor. Several growth factors induced G-CSF expression by a MEK-dependent mechanism. Inhibition of G-CSF release with a MEK inhibitor markedly reduced G-CSF production in vitro and synergized with anti-VEGF antibodies to reduce CD11b(+)Ly6G(+) neutrophil mobilization and tumor growth and led to increased survival in animal models of cancer, including a genetically engineered mouse model of pancreatic adenocarcinoma. Analysis of biopsies from pancreatic cancer patients revealed increased phospho-MEK, G-CSF, and Ets expression and enhanced neutrophil recruitment compared with normal pancreata. These results provide insights into G-CSF regulation and on the mechanism of action of MEK inhibitors and point to unique anticancer strategies.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/metabolismo , Sistema de Señalización de MAP Quinasas , Neutrófilos/citología , Proteína Proto-Oncogénica c-ets-2/metabolismo , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Animales , Sitios de Unión , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Desnudos , Ratones Transgénicos , Neoplasias/metabolismo , Neovascularización Patológica , Infiltración Neutrófila , Proteínas Tirosina Quinasas/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
11.
Mol Cancer Ther ; 11(4): 1026-35, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22302098

RESUMEN

To examine the potential of combining Bcl-2 family inhibitors with chemotherapy in ovarian cancer, we evaluated a panel of 27 ovarian cancer cell lines for response to the combination of navitoclax (formerly ABT-263) and paclitaxel or gemcitabine. The majority of cell lines exhibited a greater than additive response to either combination, as determined by the Bliss independence model, and more than 50% of the ovarian cell lines exhibited strong synergy for the navitoclax/paclitaxel combination. To identify biomarkers for tumors likely to respond to this combination, we evaluated the protein levels of intrinsic apoptosis pathway components. Bcl-x(L) seems necessary, but not sufficient, for navitoclax/paclitaxel synergy in vitro, suggesting that exclusion of patients whose tumors have low or undetectable Bcl-x(L) would enrich for patients responsive to the combination. We evaluated Bcl-x(L) levels in ovarian cancer tumor tissue from 40 patients (20 taxane responsive and 20 with poor response to taxane) and found that patients with high Bcl-x(L) were less sensitive to taxane treatment (10 of 12) Bcl-x(L) positive patients, P = 0.014). These data support the use of navitoclax in combination with taxane-based therapy in ovarian cancer patients with high levels of Bcl-x(L).


Asunto(s)
Compuestos de Anilina/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Sulfonamidas/farmacología , Proteína bcl-X/metabolismo , Compuestos de Anilina/administración & dosificación , Línea Celular Tumoral , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Sinergismo Farmacológico , Femenino , Humanos , Inmunohistoquímica , Neoplasias Ováricas/metabolismo , Paclitaxel/administración & dosificación , Paclitaxel/farmacología , Sulfonamidas/administración & dosificación , Gemcitabina
12.
Magn Reson Med ; 65(3): 889-99, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21442797

RESUMEN

Imaging of tumor microvasculature has become an important tool for studying angiogenesis and monitoring antiangiogenic therapies. Ultrasmall paramagnetic iron oxide contrast agents for indirect imaging of vasculature offer a method for quantitative measurements of vascular biomarkers such as vessel size index, blood volume, and vessel density (Q). Here, this technique is validated with direct comparisons to ex vivo micro-computed tomography angiography and histologic vessel measurements, showing significant correlations between in vivo vascular MRI measurements and ex vivo structural vessel measurements. The sensitivity of the MRI vascular parameters is also demonstrated, in combination with a multispectral analysis technique for segmenting tumor tissue to restrict the analysis to viable tumor tissue and exclude regions of necrosis. It is shown that this viable tumor segmentation increases sensitivity for detection of significant effects on blood volume and Q by two antiangiogenic therapeutics [anti-vascular endothelial growth factor (anti-VEGF) and anti-neuropilin-1] on an HM7 colorectal tumor model. Anti-vascular endothelial growth factor reduced blood volume by 36±3% (p<0.0001) and Q by 52±3% (p<0.0001) at 48 h post-treatment; the effects of anti-neuropilin-1 were roughly half as strong with a reduction in blood volume of 18±6% (p<0.05) and a reduction in Q of 33±5% (p<0.05) at 48 h post-treatment.


Asunto(s)
Neoplasias Colorrectales/diagnóstico por imagen , Neovascularización Patológica/diagnóstico por imagen , Angiografía , Animales , Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Imagen por Resonancia Magnética/métodos , Ratones , Sensibilidad y Especificidad , Tomografía Computarizada por Rayos X , Carga Tumoral/efectos de los fármacos
13.
Proc Natl Acad Sci U S A ; 107(50): 21248-55, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21081700

RESUMEN

Priming of the organ-specific premetastatic sites is thought to be an important yet incompletely understood step during metastasis. In this study, we show that the metastatic tumors we examined overexpress granulocyte-colony stimulating factor (G-CSF), which expands and mobilizes Ly6G+Ly6C+ granulocytes and facilitates their subsequent homing at distant organs even before the arrival of tumor cells. Moreover, G-CSF-mobilized Ly6G+Ly6C+ cells produce the Bv8 protein, which has been implicated in angiogenesis and mobilization of myeloid cells. Anti-G-CSF or anti-Bv8 antibodies significantly reduced lung metastasis. Transplantation of Bv8 null fetal liver cells into lethally irradiated hosts also reduced metastasis. We identified an unexpected role for Bv8: the ability to stimulate tumor cell migration through activation of one of the Bv8 receptors, prokineticin receptor (PKR)-1. Finally, we show that administration of recombinant G-CSF is sufficient to increase the numbers of Ly6G+Ly6C+ cells in organ-specific metastatic sites and results in enhanced metastatic ability of several tumors.


Asunto(s)
Antígenos Ly/inmunología , Factor Estimulante de Colonias de Granulocitos/farmacología , Granulocitos/efectos de los fármacos , Granulocitos/inmunología , Neoplasias Pulmonares/patología , Metástasis de la Neoplasia , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular , Femenino , Perfilación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos/genética , Granulocitos/citología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones SCID , Análisis por Micromatrices , Trasplante de Neoplasias , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
14.
Magn Reson Med ; 63(6): 1637-47, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20512867

RESUMEN

Imaging of tumor microvasculature has become an important tool for studying angiogenesis and monitoring antiangiogenic therapies. Ultrasmall paramagnetic iron oxide contrast agents for indirect imaging of vasculature offer a method for quantitative measurements of vascular biomarkers such as vessel size index, blood volume, and vessel density. Here, this technique is validated with direct comparisons to ex vivo micro-CT angiography and histologic vessel measurements, showing significant correlations between in vivo vascular MRI measurements and ex vivo structural vessel measurements. The sensitivity of the MRI vascular parameters is also demonstrated, in combination with a multispectral analysis technique for segmenting tumor tissue to restrict the analysis to viable tumor tissue and exclude regions of necrosis. It is shown that this viable tumor segmentation increases sensitivity for detection of significant effects on blood volume and vessel density by two antiangiogenic therapeutics (anti-VEGF and anti-neuropilin-1) on an HM7 colorectal tumor model. Anti-VEGF reduced blood volume by 36 +/- 3% (P < 0.0001) and vessel density by 52 +/- 3% (P < 0.0001) at 48 h posttreatment; the effects of anti-neuropilin-1 were roughly half as strong with a reduction in blood volume of 18 +/- 6% (P < 0.05) and a reduction in vessel density of 33 +/- 5% (P < 0.05) at 48 h posttreatment.


Asunto(s)
Angiografía , Neoplasias Colorrectales/diagnóstico por imagen , Imagen por Resonancia Magnética , Neovascularización Patológica/diagnóstico por imagen , Tomografía Computarizada por Rayos X , Animales , Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Imagen por Resonancia Magnética/métodos , Ratones , Sensibilidad y Especificidad , Carga Tumoral/efectos de los fármacos
15.
Clin Cancer Res ; 15(21): 6674-82, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19861458

RESUMEN

PURPOSE: Little is known concerning the onset, duration, and magnitude of direct therapeutic effects of anti-vascular endothelial growth factor (VEGF) therapies. Such knowledge would help guide the rational development of targeted therapeutics from bench to bedside and optimize use of imaging technologies that quantify tumor function in early-phase clinical trials. EXPERIMENTAL DESIGN: Preclinical studies were done using ex vivo microcomputed tomography and in vivo ultrasound imaging to characterize tumor vasculature in a human HM-7 colorectal xenograft model treated with the anti-VEGF antibody G6-31. Clinical evaluation was by quantitative magnetic resonance imaging in 10 patients with metastatic colorectal cancer treated with bevacizumab. RESULTS: Microcomputed tomography experiments showed reduction in perfused vessels within 24 to 48 h of G6-31 drug administration (P

Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/tratamiento farmacológico , Diagnóstico por Imagen , Factor A de Crecimiento Endotelial Vascular/inmunología , Adolescente , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Bevacizumab , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Ratones , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Magn Reson Imaging ; 28(4): 996-1004, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18821600

RESUMEN

PURPOSE: To develop magnetic resonace imaging (MRI) methods for functional assessment of arteriogenesis in a murine model of peripheral artery disease to quantify the influences of vascular endothelial growth factor (VEGF), age, and atherosclerosis. MATERIALS AND METHODS: Reactive hyperemia (RH), which was induced using a device designed for remote and transient occlusion of the aorta and vena cava, was measured by blood-oxygen-level-dependent MRI. Twenty-eight days after femoral artery ligation, peak height (PH) and time to peak (TTP) of the RH response was compared with sham-operated animals in 10-week-old C57Bl6, 9-month-old C57Bl6, and 9-month-old Ldlr(-/-)Apobec(-/-) mice. The contribution of VEGF to functional recovery was assessed in young mice. Angiogenesis was quantified using an anti-PECAM1 radioimmunoassay. RESULTS: In young animals, angiogenesis was maximal 7 days after ligation, whereas functional recovery took 28 days. Inhibition of VEGF eliminated the angiogenesis seen at 7 days and reduced RH (PH, P < 0.05). At day 28, RH was altered in old (TTP, P < 0.05) and atherosclerotic (PH, P < 0.05; TTP, P < 0.05) animals. RH was different in young, old, and atherosclerotic sham animals. Old and atherosclerotic mice showed reduced angiogenesis. CONCLUSION: The method presented herein can provide a sensitive assay for the functional assessment of arteriogenesis and highlights the contribution of VEGF, age, and atherosclerosis to this process.


Asunto(s)
Aterosclerosis/fisiopatología , Hiperemia/fisiopatología , Imagen por Resonancia Magnética/métodos , Neovascularización Fisiológica/efectos de los fármacos , Enfermedades Vasculares Periféricas/fisiopatología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factores de Edad , Análisis de Varianza , Animales , Aterosclerosis/metabolismo , Modelos Animales de Enfermedad , Miembro Posterior , Hiperemia/metabolismo , Ratones , Enfermedades Vasculares Periféricas/metabolismo , Radioinmunoensayo , Recuperación de la Función , Factores de Riesgo , Factores de Tiempo
17.
Clin Cancer Res ; 14(1): 249-58, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18172277

RESUMEN

PURPOSE: Multiple endocrine neoplasia type 1 (MEN1) is defined clinically by the combined occurrence of multiple tumors, typically of the parathyroid glands, pancreatic islet cells, and anterior pituitary gland. A mouse model with a heterozygous deletion of the Men1 gene recapitulates the tumorigenesis of MEN1. We wished to determine the role of vascular endothelial growth factor (VEGF)-A in the vascularization and growth of MEN1-associated tumors, with an emphasis on pituitary adenomas. EXPERIMENTAL DESIGN: To investigate whether tumor growth in Men1(+/-) mice is mediated by VEGF-A dependent angiogenesis, we carried out a monotherapy with the anti-VEGF-A monoclonal antibody (mAb) G6-31. We evaluated tumor growth by magnetic resonance imaging and assessed vascular density in tissue sections. We also measured hormone levels in the serum. RESULTS: During the treatment with mAb G6-31, a significant inhibition of the pituitary adenoma growth was observed, leading to an increased mean tumor doubling-free survival compared with mice treated with a control antibody. Similarly, the growth of s.c. pituitary adenoma transplants was effectively inhibited by administration of anti-VEGF-A mAb. Serum prolactin was lowered by mAb G6-31 treatment but not by control antibody, potentially providing a new therapeutic approach for treating the hormonal excess in MEN1 patients. Additionally, the vascular density in pancreatic islet tumors was significantly reduced by the treatment. CONCLUSIONS: These results suggest that VEGF-A blockade may represent a nonsurgical treatment for benign tumors of the endocrine system.


Asunto(s)
Adenoma/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/patología , Neoplasias Hipofisarias/patología , Prolactina/sangre , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Adenoma/sangre , Adenoma/patología , Animales , Anticuerpos Monoclonales/farmacología , Modelos Animales de Enfermedad , Inmunohistoquímica , Imagen por Resonancia Magnética , Ratones , Ratones Mutantes , Ratones Desnudos , Neoplasia Endocrina Múltiple Tipo 1/sangre , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neovascularización Patológica , Neoplasias Hipofisarias/sangre , Neoplasias Hipofisarias/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos
18.
Nature ; 450(7171): 825-31, 2007 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-18064003

RESUMEN

Bone-marrow-derived cells facilitate tumour angiogenesis, but the molecular mechanisms of this facilitation are incompletely understood. We have previously shown that the related EG-VEGF and Bv8 proteins, also known as prokineticin 1 (Prok1) and prokineticin 2 (Prok2), promote both tissue-specific angiogenesis and haematopoietic cell mobilization. Unlike EG-VEGF, Bv8 is expressed in the bone marrow. Here we show that implantation of tumour cells in mice resulted in upregulation of Bv8 in CD11b+Gr1+ myeloid cells. We identified granulocyte colony-stimulating factor as a major positive regulator of Bv8 expression. Anti-Bv8 antibodies reduced CD11b+Gr1+ cell mobilization elicited by granulocyte colony-stimulating factor. Adenoviral delivery of Bv8 into tumours was shown to promote angiogenesis. Anti-Bv8 antibodies inhibited growth of several tumours in mice and suppressed angiogenesis. Anti-Bv8 treatment also reduced CD11b+Gr1+ cells, both in peripheral blood and in tumours. The effects of anti-Bv8 antibodies were additive to those of anti-Vegf antibodies or cytotoxic chemotherapy. Thus, Bv8 modulates mobilization of CD11b+Gr1+ cells from the bone marrow during tumour development and also promotes angiogenesis locally.


Asunto(s)
Hormonas Gastrointestinales/metabolismo , Células Mieloides/metabolismo , Neoplasias/irrigación sanguínea , Neovascularización Patológica , Neuropéptidos/metabolismo , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Antineoplásicos/farmacología , División Celular/efectos de los fármacos , Línea Celular Tumoral , Hormonas Gastrointestinales/antagonistas & inhibidores , Hormonas Gastrointestinales/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Ratones , Ratones Desnudos , Células Mieloides/efectos de los fármacos , Trasplante de Neoplasias , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico , Neuropéptidos/antagonistas & inhibidores , Neuropéptidos/inmunología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
19.
Proc Natl Acad Sci U S A ; 104(25): 10625-30, 2007 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-17553957

RESUMEN

Anti-VEGF-A monoclonal antibodies, in combination with chemotherapy, result in a survival benefit in patients with metastatic colorectal and non-small cell lung cancer, but little is known regarding the impact of anti-VEGF-A therapy on benign or premalignant tumors. The Apc+/min mice have been widely used as a model recapitulating early intestinal adenoma formation. To investigate whether tumor growth in Apc+/min mice is mediated by VEGF-A-dependent angiogenesis, we used two independent approaches to inhibit VEGF-A: monotherapy with a monoclonal antibody (Mab) targeting VEGF-A and genetic deletion of VEGF-A selectively in intestinal epithelial cells. Short-term (3 or 6 weeks) treatment with anti-VEGF-A Mab G6-31 resulted in a nearly complete suppression of adenoma growth throughout the small intestine. Growth inhibition by Mab G6-31 was associated with a decrease in vascular density. Long-term (up to 52 weeks) treatment with Mab G6-31 led to a substantial increase in median survival. Deletion of VEGF-A in intestinal epithelial cells of Apc+/min mice yielded a significant inhibition of tumor growth, albeit of lesser magnitude than that resulting from Mab G6-31 administration. These results establish that inhibition of VEGF-A signaling is sufficient for tumor growth cessation and confers a long-term survival benefit in an intestinal adenoma model. Therefore, VEGF-A inhibition may be a previously uncharacterized strategy for the prevention of the angiogenic switch and growth in intestinal adenomas.


Asunto(s)
Genes APC/fisiología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Adenoma/irrigación sanguínea , Adenoma/genética , Adenoma/inmunología , Adenoma/terapia , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Eliminación de Gen , Hibridación in Situ , Neoplasias Intestinales/irrigación sanguínea , Neoplasias Intestinales/genética , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/terapia , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología , Análisis de Supervivencia , Factores de Tiempo
20.
J Magn Reson Imaging ; 24(5): 1124-32, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17029233

RESUMEN

PURPOSE: To quantify spontaneous and therapeutic arteriogenesis in vivo in a murine model of peripheral arterial disease using magnetic resonance angiography. MATERIALS AND METHODS: Male, 8-12-week-old, C57/BL6 mice underwent femoral artery ligation; 21 days later, 2 mg/kg recombinant murine VEGF165, formulated for slow release, was injected into the ipsilateral gastrocnemius. The spontaneous (following ligation) and therapeutic (following vascular endothelial growth factor (VEGF)) formation of collateral vessels was quantified using 3D magnetic resonance angiography on a small-bore 4.7T system. Therapeutically induced angiogenesis and blood flow were quantified using an in situ anti-platelet endothelial cell adhesion molecule (PECAM) 1 radioimmunoassay and radiolabeled microsphere deposition, respectively. RESULTS: Spontaneous arteriogenesis was visible in all animals five days after ligation. VEGF treatment doubled the arteriogenic response five days after treatment compared to vehicle (cross-sectional area of vessels: 0.96 vs. 0.46 mm2, P<0.01). VEGF also induced angiogenesis (PECAM1 levels 191% of vehicle, P<0.05) and increased blood flow specific to the injection site (57 vs. 7 mL/minute/100 g, P<0.05). CONCLUSION: The presented methodology allowed in vivo quantification of spontaneous arteriogenesis in a murine model of peripheral arterial disease and demonstrated that therapeutic enlargement of collateral vessels is possible with VEGF.


Asunto(s)
Velocidad del Flujo Sanguíneo/efectos de los fármacos , Modelos Animales de Enfermedad , Angiografía por Resonancia Magnética/métodos , Neovascularización Fisiológica/efectos de los fármacos , Enfermedades Vasculares Periféricas/tratamiento farmacológico , Enfermedades Vasculares Periféricas/patología , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Animales , Circulación Colateral/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Pronóstico , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA