Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Life Sci ; 291: 120240, 2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-34942164

RESUMEN

AIMS: In this study, we used a near-infrared laser (NIR) to increase the potency of silver nanoparticles (AgNPs) to develop a novel, less invasive, and simple photothermal therapy technique for benign prostate hyperplasia (BPH). MATERIALS AND METHODS: The shape, particle size, and zeta-potential of polyvinylpyrrolidone coated-AgNPs (PVP-AgNPs) were determined using transmission electron microscopy (TEM), Zeta-potential, and Particle size analyzer (ELSZ). To induce BPH, thirty-six male Sprague-Dawley (SD) rats were given intramuscular (i.m) injections of testosterone propionate (TP) at 5 mg/kg body weight (b.w)/day suspended in 0.1 ml of olive oil for 14 days. Photothermal therapy with AgNPs-NIR for 14 days was carried out. Prostate size, prostate index (PI), dihydrotestosterone (DHT), prostate-specific antigen (PSA), gross, hepatic, and renal toxicity, as well as antioxidant activity, apoptosis, and angiogenesis markers in prostatic tissues were measured. Histological examinations of prostates and biocompatibility of NIR-AgNPs on vital organs were also performed. KEY FINDINGS: The aggregated spherical AgNPs with a mean size of 50-90 nm and a Zeta potential of -53.22 mV displayed high effectiveness in the NIR (532 nm-1 W) region by decreasing prostate size, PI, DHT, and PSA in BPH rats with no signs of gross, hepatic, or renal damage. As compared to alternative therapies, hyperthermia therapy increased antioxidant activities, induced apoptosis, inhibited angiogenesis, reduced histological alterations in the prostates of BPH rats, and improved biocompatibility of the vital organs. SIGNIFICANCE: The current study demonstrated the effectiveness of plasmonic AgNPs photothermal therapy in the treatment of BPH.


Asunto(s)
Nanopartículas del Metal/uso terapéutico , Fototerapia/métodos , Hiperplasia Prostática/tratamiento farmacológico , Animales , Antioxidantes/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Dihidrotestosterona/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Finasterida/farmacología , Hiperplasia/patología , Masculino , Nanopartículas del Metal/química , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/fisiopatología , Extractos Vegetales/farmacología , Próstata/patología , Hiperplasia Prostática/fisiopatología , Ratas , Ratas Sprague-Dawley , Plata/química , Resonancia por Plasmón de Superficie/métodos , Testosterona/efectos adversos , Propionato de Testosterona/uso terapéutico
2.
Chin J Nat Med ; 18(9): 696-703, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32928513

RESUMEN

Hypoxia is a prominent feature of tumors. Hypoxia-inducible factor-1α (HIF-1α), a major subunit of HIF-1, is overexpressed in hypoxic tumor tissues and activates the transcription of many oncogenes. Accumulating evidence has demonstrated that HIF-1α promotes tumor angiogenesis, metastasis, metabolism, and immune evasion. Natural products are an important source of antitumor drugs and numerous studies have highlighted the crucial role of these agents in modulating HIF-1α. The present review describes the role of HIF-1α in tumor progression, summarizes natural products used as HIF-1α inhibitors, and discusses the potential of developing natural products as HIF-1α inhibitors for the treatment of cancer.


Asunto(s)
Antineoplásicos/farmacología , Productos Biológicos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias/fisiopatología , Antineoplásicos/química , Línea Celular Tumoral , Humanos , Neovascularización Patológica/fisiopatología
3.
Int J Pharm ; 585: 119556, 2020 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-32574684

RESUMEN

In recent years, due to the effective drug delivery and preciseness of tumor sites or microenvironment, the targeted drug delivery approaches have gained ample attention for tumor metastasis therapy. The conventional treatment approaches for metastasis therapy have reported with immense adverse effects because they exhibited maximum probability of killing the carcinogenic cells along with healthy cells. The tumor vasculature, comprising of vasculogenic impressions and angiogenesis, greatly depends upon the growth and metastasis in the tumors. Therefore, various nanocarriers-based delivery approaches for targeting to tumor vasculature have been attempted as efficient and potential approaches for the treatment of tumor metastasis and the associated lesions. Furthermore, the targeted drug delivery approaches have found to be most apt way to overcome from all the limitations and adverse effects associated with the conventional therapies. In this review, various approaches for efficient targeting of pharmacologically active chemotherapeutics against tumor metastasis with the cohesive objectives of prognosis, tracking and therapy are summarized.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Portadores de Fármacos/química , Humanos , Lípidos/química , Nanopartículas del Metal/química , Metástasis de la Neoplasia , Neovascularización Patológica/fisiopatología , Péptidos/fisiología , Fototerapia/métodos , Polímeros/química , ARN/fisiología , Microambiente Tumoral/fisiología
4.
Cells ; 8(6)2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31151284

RESUMEN

Background: Angiogenesis inhibitors (AIs) have become established as an effective cancer treatment. Whereas their interactions with antineoplastic drugs have extensively been investigated, little is known of the effect of their co-administration with nutraceuticals/dietary supplements (N/DSs), which are often self-prescribed. N/DSs comprise a wide range of products such as herbs, nutrients, vitamins, minerals, and probiotics. Assessment of their interactions with cancer drugs, particularly AIs, is hampered by the difficulty of gauging the amount of active substances patients actually take. Moreover, there is no agreement on which approach should be used to determine which N/DSs are most likely to influence AI treatment efficacy. We present a comprehensive review of the metabolic routes of the major AIs and their possible interactions with N/DSs. Methods: The PubMed and Cochrane databases were searched for papers describing the metabolic routes of the main AIs and N/DSs. Results: Data from the 133 studies thus identified were used to compile a diagnostic table reporting known and expected AI-N/DS interactions based on their metabolization pathways. AIs and N/DSs sharing the cytochrome P450 pathway are at risk of negative interactions. Conclusions: Recent advances in pharmacogenetics offer exceptional opportunities to identify prognostic and predictive markers to enhance the efficacy of individualized AI treatments. The table provides a guide to genotyping patients who are due to receive AIs and is a promising tool to prevent occult AI-N/DS interactions in poor metabolizers. N/DS use by cancer patients receiving AIs is a topical problem requiring urgent attention from the scientific community.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Suplementos Dietéticos , Interacciones Farmacológicas , Farmacogenética , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Humanos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/fisiopatología , Factor A de Crecimiento Endotelial Vascular/genética
5.
Food Funct ; 10(5): 2605-2617, 2019 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-31020299

RESUMEN

The rhizome of Alpinia officinarum Hance, a popular spice used as a condiment in China and Europe, has various reported bioactivities, including anticancer, anti-inflammatory and antioxidant effects. However, its anti-angiogenic activity has not previously been reported. In this study, a diarylheptanoid was isolated from Alpinia officinarum and identified as 1-phenyl-7-(4-hydroxy-3-methoxyphenyl)-4E-en-3-heptanone (PHMH). We demonstrated that PHMH exerts anti-angiogenic activity both in vitro and in vivo. PHMH inhibited vascular endothelial growth factor (VEGF)-induced viability, migration, invasion and tube formation in human umbilical vein endothelial cells (HUVECs) in vitro, and also suppressed VEGF-induced sprout formation of rat aorta ex vivo. Furthermore, PHMH was found to block VEGF-induced vessel formation in mice and suppress angiogenesis in both zebrafish and chorioallantoic membrane models. Mechanistic studies indicated that PHMH inhibited VEGF-induced VEGF receptor-2 (VEGFR-2) auto-phosphorylation and resulted in the blockage of VEGFR-2-mediated signaling cascades in HUVECs, including the Akt/mTOR, ERK1/2, and FAK pathways. Our findings provide new insights into the potential application of PHMH as a therapeutic agent for anti-angiogenesis.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Diarilheptanoides/administración & dosificación , Medicamentos Herbarios Chinos/administración & dosificación , Neovascularización Patológica/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Alpinia , Inhibidores de la Angiogénesis/química , Animales , Movimiento Celular/efectos de los fármacos , China , Diarilheptanoides/química , Medicamentos Herbarios Chinos/química , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Pez Cebra
6.
Arterioscler Thromb Vasc Biol ; 39(4): 635-646, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30786744

RESUMEN

Nanoparticles promise to advance strategies to treat vascular disease. Since being harnessed by the cancer field to deliver safer and more effective chemotherapeutics, nanoparticles have been translated into applications for cardiovascular disease. Systemic exposure and drug-drug interactions remain a concern for nearly all cardiovascular therapies, including statins, antithrombotic, and thrombolytic agents. Moreover, off-target effects and poor bioavailability have limited the development of completely new approaches to treat vascular disease. Through the rational design of nanoparticles, nano-based delivery systems enable more efficient delivery of a drug to its therapeutic target or even directly to the diseased site, overcoming biological barriers and enhancing a drug's therapeutic index. In addition, advances in molecular imaging have led to the development of theranostic nanoparticles that may simultaneously act as carriers of both therapeutic and imaging payloads. The following is a summary of nanoparticle therapy for atherosclerosis, thrombosis, and restenosis and an overview of recent major advances in the targeted treatment of vascular disease.


Asunto(s)
Fármacos Cardiovasculares/administración & dosificación , Portadores de Fármacos/administración & dosificación , Nanopartículas/administración & dosificación , Enfermedades Vasculares/tratamiento farmacológico , Animales , Quimiotaxis de Leucocito/efectos de los fármacos , Colesterol/metabolismo , Evaluación Preclínica de Medicamentos , Predicción , Humanos , Inflamación/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Ratones , Neointima/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/fisiopatología , Placa Aterosclerótica/fisiopatología , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Trombosis/tratamiento farmacológico
7.
J Agric Food Chem ; 67(10): 2856-2864, 2019 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-30785275

RESUMEN

Among the various treatments, induction of synoviocyte apoptosis by natural products during a rheumatoid arthritis (RA) pathological condition can be considered to have vast potential. However, it is unclear that liquiritin, a kind of natural flavonoid extracted from the roots of Glycyrrhiza uralensis, induced the apoptosis of the synovial membrane and its molecular mechanism. In this study, interleukin-1ß (IL-1ß)-RA-FLS cells were incubated with different concentrations of liquiritin. An MTT assay, Hoechst 33342 staining, JC-1 staining, and Western blot were used to check the viability, cell apoptosis, mitochondrial membrane potential changes, and the expression of related proteins, respectively. In vivo, a TUNEL assay and HE staining of tissue were used for histopathological evaluation. Our results showed that liquiritin significantly inhibited the proliferation of IL-1ß-induced-RA-FLS, promoted nuclear DNA fragmentation, and changed the mitochondrial membrane potential to accelerate cell apoptosis. Liquiritin downregulated the ratio of Bcl-2/Bax and inhibited the VEGF expression and phosphorylation of JNK and P38. Moreover, liquiritin improved the clinical score of rheumatism, inflammatory infiltration, and angiogenesis and induced apoptosis of the synovial tissue in vivo. Hence, liquiritin ameliorates RA by reducing inflammation, blocking MAPK signaling, and restraining angiogenesis.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Medicamentos Herbarios Chinos/administración & dosificación , Flavanonas/administración & dosificación , Glucósidos/administración & dosificación , Glycyrrhiza uralensis/química , Neovascularización Patológica/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Artritis Reumatoide/genética , Artritis Reumatoide/fisiopatología , Proliferación Celular/efectos de los fármacos , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Neovascularización Patológica/genética , Neovascularización Patológica/inmunología , Neovascularización Patológica/fisiopatología , Fosforilación/efectos de los fármacos , Ratas , Ratas Wistar , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
8.
Clin Cancer Res ; 25(3): 912-920, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30274981

RESUMEN

Hepatocellular carcinoma (HCC) accounts for about 90% of all primary liver cancers and is the second leading cause of cancer-related deaths worldwide. The hypervascular nature of most HCC tumors underlines the importance of angiogenesis in the pathobiology of these tumors. Several angiogenic pathways have been identified as being dysregulated in HCC, suggesting they may be involved in the development and pathogenesis of HCC. These data provide practical targets for systemic treatments such as those targeting the vascular endothelial growth factor receptor and its ligand. However, the clinical relevance of other more recently identified angiogenic pathways in HCC pathogenesis or treatment remains unclear. Research into molecular profiles and validation of prognostic or predictive biomarkers will be required to identify the patient subsets most likely to experience meaningful benefit from this important class of agents.


Asunto(s)
Angiostatinas/uso terapéutico , Carcinoma Hepatocelular/irrigación sanguínea , Neoplasias Hepáticas/irrigación sanguínea , Neovascularización Patológica/tratamiento farmacológico , Sorafenib/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Neovascularización Patológica/patología , Neovascularización Patológica/fisiopatología , Pronóstico , Inhibidores de Proteínas Quinasas/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Int J Low Extrem Wounds ; 17(4): 301-303, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30501554

RESUMEN

In recent years, wound repair, especially chronic and refractory wounds, is still a growing worldwide health problem that concerns the physical and mental health of patients and the productivity of society as a whole. In many developed countries and developing countries, such as China, rapid economic growth, negative lifestyle changes (such as increased dietary fat intake and reduced physical activity), and increasingly serious aging problems have led to more and more refractory wounds caused by chronic diseases, such as diabetes and microvascular diseases. There is a clear and orderly stage in the process of wound healing, and the malfunction of any step will lead to poor healing effect. There are many causes of refractory wounds, among which the poor vascular network is closely related. This review focuses on the role and improvement of the microvasculature in wound repair. In addition, the future developmental trend of wound regenerative therapy (active factors, [stem] cells, tissue-induced biomaterials, gene therapy, etc) is also considered.


Asunto(s)
Terapia Biológica/métodos , Neovascularización Patológica/patología , Cicatrización de Heridas/fisiología , Heridas y Lesiones/terapia , Factores de Edad , Anciano , Anciano de 80 o más Años , Envejecimiento/fisiología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , China , Enfermedad Crónica , Terapia Combinada , Femenino , Terapia Genética/métodos , Humanos , Masculino , Neovascularización Patológica/epidemiología , Neovascularización Patológica/fisiopatología , Pronóstico , Medición de Riesgo , Índice de Severidad de la Enfermedad , Factores Sexuales , Heridas y Lesiones/fisiopatología
10.
J Hematol Oncol ; 10(1): 74, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28335793

RESUMEN

BACKGROUND: Current angiogenic therapies for cancers and cardiovascular diseases have not yet achieved expected benefits, which reflects the need for improved understanding of angiogenesis. In this study, we focused on solving the problem of whether tissues have different angiogenic potentials (APs) in physiological conditions and how angiogenesis is regulated in various disease conditions. METHODS: In healthy and diseased human and mouse tissues, we profiled the expression of 163 angiogenic genes, including transcription regulators (TRs), growth factors and receptors (GF/Rs), cytokines and chemokines (C/Cs), and proteases and inhibitors (P/Is). TRs were categorized as inflammatory, homeostatic, and endothelial cell-specific TRs, and C/Cs were categorized as pro-angiogenic, anti-angiogenic, and bi-functional C/Cs. RESULTS: We made the following findings: (1) the human heart, muscle, eye, pancreas, and lymph node are among the tissues with the highest APs; (2) tissues with high APs have more active angiogenic pathways and angiogenic C/C responses; (3) inflammatory TRs dominate regulation of all angiogenic C/Cs; homeostatic TRs regulate all to a lower extent, while endothelial cell-specific TRs mainly regulate pro-angiogenic and bi-functional C/Cs; (4) tissue AP is positively correlated with the expression of oxygen sensors PHD2 and HIF1B, VEGF pathway gene VEGFB, and stem cell gene SOX2; (5) cancers of the digestive system tend to have increased angiogenesis dominated by endothelial cell-specific pro-angiogenic pathways, while lung cancer and prostate cancer have significantly decreased angiogenesis; and (6) endothelial cell-specific pro-angiogenic pathways are significantly increased in thrombus-derived leukocytes in patients with acute coronary artery disease. CONCLUSIONS: Our results demonstrate that thrombus-derived leukocytes express more endothelial cell-specific angiogenic markers to directly promote angiogenesis after myocardial infarction and that certain solid tumors may be more sensitive to anti-angiogenic therapies than others.


Asunto(s)
Síndrome Coronario Agudo/fisiopatología , Transdiferenciación Celular , Células Endoteliales , Leucocitos/patología , Neovascularización Fisiológica , Trombosis/patología , Proteínas Angiogénicas/genética , Animales , Biomarcadores , Minería de Datos , Humanos , Ratones , Infarto del Miocardio/fisiopatología , Neoplasias/irrigación sanguínea , Neoplasias/fisiopatología , Neovascularización Patológica/fisiopatología , Transcriptoma
11.
Zhonghua Wei Chang Wai Ke Za Zhi ; 20(1): 84-89, 2017 Jan 25.
Artículo en Chino | MEDLINE | ID: mdl-28105626

RESUMEN

OBJECTIVE: To investigate the effects of polyunsaturated fatty acids (PUFA) ω-3 and ω-6, and their middle metabolites PGE2 and PGE3 on angiogenesis formation of gastric cancer, and to explore associated mechanism. METHODS: The effects of ω-3, ω-6, PGE2, PGE3 on the proliferation and migration of human umbilical vein endothelial cell (HUVEC) were measured by proliferation and migration assay respectively. The angiogenesis assay in vivo was used to measure the effects of ω-3, ω-6, PGE2 and PGE3 on neovascularization. In all the assays, groups without ω-3, ω-6, PGE2 and PGE3 were designed as the control. RESULTS: With the increased concentration of ω-6 from 1 µmol/L to 10 µmol/L, the proliferation ability of HUVECs enhanced, and the number of migration cells also increased from 28.2±3.0 to 32.8±2.1, which was higher than control group (21.2±3.2) respectively (both P<0.05). With the increased concentration of ω-3 from 1 µmol/L to 10 µmol/L, the proliferation ability of HUVECs was inhibited, and the number of migration cells decreased from 15.8±2.0 to 11.0±2.1, which was lower than control group (22.1±3.0) respectively (both P<0.05). In the angiogenesis assay, compared with control group (standard number: 43 721±4 654), the angiogenesis ability of HUVECs was significantly enhanced by ω-6 in concentration-dependent manner (1 µmol/L group: 63 238±4 795, 10 µmol/L group: 78 166±6 123, all P<0.01). Meanwhile, with the increased concentration of ω-3 from 1 µmol/L to 10 µmol/L, the angiogenesis ability was significantly decreased from 30 129±3 102 to 20 012±1 541(all P<0.01). The proliferation and migration ability of HUVECs were significantly promoted by ω-6 metabolites PGE2 (P<0.05) in a concentration-dependent manner. In contrast, ω-3 metabolites PGE3 significantly inhibited the proliferation and migration ability of HUVECs in a concentration-dependent manner (all P<0.05). After rofecoxib (a COX-2 specific inhibitor) inhibited the expression of COX-2, the expression level of PGE2 was significantly decreased in a dose-dependent manner. In co-culture system, whose gastric cancer cells expressed positive COX-2, ω-6 could increase angiogenesis of gastric cancer cells(P<0.01), but ω-3 could inhibit such angiogenesis(P<0.01). In co-culture system, whose gastric cancer cells did not express COX-2, ω-3 could inhibit the angiogenesis of gastric cancer cells (P<0.05), but ω-6 had no effect on angiogenesis. CONCLUSIONS: The PUFA ω-6 can enhance the angiogenesis via the promotion of proliferation and migration of HUVECs, and COX-2 and PGE2 may play an important role in this process, whereas, the ω-3 can inhibit the angiogenesis through its middle metabolites PGE3 to inhibit the proliferation and migration of HUVECs. Results of this experiment may provide a new approach to inhibit and prevent the spread of gastric cancer.


Asunto(s)
Alprostadil/análogos & derivados , Inductores de la Angiogénesis/farmacología , Línea Celular Tumoral/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dinoprostona/farmacología , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Omega-6/farmacología , Ácidos Grasos Insaturados/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Neovascularización Patológica/fisiopatología , Alprostadil/farmacología , Inductores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/farmacología , Recuento de Células/métodos , Línea Celular Tumoral/fisiología , Ensayos de Migración Celular , Técnicas de Cocultivo , Ciclooxigenasa 2/farmacología , Dinoprostona/metabolismo , Ácidos Grasos Omega-6/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Lactonas/farmacología , Neoplasias Gástricas/fisiopatología , Sulfonas/farmacología
12.
Clin Cancer Res ; 23(3): 666-676, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27769988

RESUMEN

PURPOSE: Bone marrow-derived progenitor cells, including VEGFR2+ endothelial progenitor cells (EPCs) and copper-dependent pathways, model the tumor microenvironment. We hypothesized that copper depletion using tetrathiomolybdate would reduce EPCs in high risk for patients with breast cancer who have relapsed. We investigated the effect of tetrathiomolybdate on the tumor microenvironment in preclinical models. EXPERIMENTAL DESIGN: Patients with stage II triple-negative breast cancer (TNBC), stage III and stage IV without any evidence of disease (NED), received oral tetrathiomolybdate to maintain ceruloplasmin (Cp) between 8 and 17 mg/dL for 2 years or until relapse. Endpoints were effect on EPCs and other biomarkers, safety, event-free (EFS), and overall survival (OS). For laboratory studies, MDA-LM2-luciferase cells were implanted into CB17-SCID mice and treated with tetrathiomolybdate or water. Tumor progression was quantified by bioluminescence imaging (BLI), copper depletion status by Cp oxidase levels, lysyl oxidase (LOX) activity by ELISA, and collagen deposition. RESULTS: Seventy-five patients enrolled; 51 patients completed 2 years (1,396 cycles). Most common grade 3/4 toxicity was neutropenia (3.7%). Lower Cp levels correlated with reduced EPCs (P = 0.002) and LOXL-2 (P < 0.001). Two-year EFS for patients with stage II-III and stage IV NED was 91% and 67%, respectively. For patients with TNBC, EFS was 90% (adjuvant patients) and 69% (stage IV NED patients) at a median follow-up of 6.3 years, respectively. In preclinical models, tetrathiomolybdate decreased metastases to lungs (P = 0.04), LOX activity (P = 0.03), and collagen crosslinking (P = 0.012). CONCLUSIONS: Tetrathiomolybdate is safe, well tolerated, and affects copper-dependent components of the tumor microenvironment. Biomarker-driven clinical trials in high risk for patients with recurrent breast cancer are warranted. Clin Cancer Res; 23(3); 666-76. ©2016 AACR.


Asunto(s)
Adenocarcinoma/secundario , Neoplasias de la Mama/tratamiento farmacológico , Quelantes/uso terapéutico , Cobre/metabolismo , Células Progenitoras Endoteliales/efectos de los fármacos , Neoplasias Pulmonares/secundario , Molibdeno/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/prevención & control , Aminoácido Oxidorreductasas/sangre , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Ceruloplasmina/análisis , Quelantes/farmacología , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Células Progenitoras Endoteliales/fisiología , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Pulmonares/prevención & control , Ratones SCID , Molibdeno/farmacología , Proteínas de Neoplasias/sangre , Neovascularización Patológica/fisiopatología , Neovascularización Patológica/prevención & control , Neutropenia/inducido químicamente , Riesgo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Strahlenther Onkol ; 192(3): 174-81, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26501141

RESUMEN

BACKGROUND: Inspiratory hyperoxia under hyperbaric conditions has been shown to effectively reduce tumor hypoxia and to improve radiosensitivity. However, applying irradiation (RT) under hyperbaric conditions is technically difficult in the clinical setting since RT after decompression may be effective only if tumor pO2 remains elevated for a certain period of time. The aim of the present study was to analyze the time course of tumor oxygenation and perfusion during and after hyperbaric hyperoxia. MATERIALS AND METHODS: Tumor oxygenation, red blood cell (RBC) flux for perfusion monitoring, and vascular resistance were assessed continuously in experimental rat DS-sarcomas by polarographic catheter electrodes and laser Doppler flowmetry at 1 and 2 atm (bar) of environmental pressure during breathing of pure O2 or carbogen (95 % O2 + 5 % CO2). RESULTS: During room air breathing, the tumor pO2 followed very rapidly within a few minutes the change of the ambient pressure during compression or decompression. With O2 breathing under hyperbaric conditions, the tumor pO2 increased more than expected based on the rise of the environmental pressure, although the time course was comparably rapid. Breathing carbogen, the tumor pO2 followed with a slight delay of the pressure change, and within 10 min after decompression the baseline values were reached again. RBC flux increased during carbogen breathing but remained almost constant with pure O2, indicating a vasodilation (decrease in vascular resistance) with carbogen but a vasoconstriction (increase in vascular resistance) with O2 during hyperbaric conditions. CONCLUSION: Since the tumor pO2 directly followed the environmental pressure, teletherapy after hyperbaric conditions does not seem to be promising as the pO2 reaches baseline values again within 5-10 min after decompression.


Asunto(s)
Oxigenoterapia Hiperbárica/métodos , Inhalación , Neovascularización Patológica/fisiopatología , Consumo de Oxígeno , Oxígeno/metabolismo , Sarcoma/fisiopatología , Animales , Velocidad del Flujo Sanguíneo , Masculino , Tasa de Depuración Metabólica , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley , Sarcoma/irrigación sanguínea
14.
J Tradit Chin Med ; 35(2): 160-7, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25975048

RESUMEN

OBJECTIVE: To evaluated the effect of calycosin on left ventricular ejection fraction and angiogenesis. METHODS: Adult male Sprague-Dawley rats were randomly assigned into calycosin-treated groups (0.5, 1, 2, and 4 mg/kg qd), a dimethyl sulfoxide (DMSO), or a sham-operated control group. The myocardial ischaemia (MI) model was intraperitoneally administered calycosin for 28 days. The survival rates and left ventricular ejection fractions (LVEF) were compared between groups. The expression levels of vascular endothelial growth factor (VEGF) and cluster of differentiation 31 (CD31) in ischaemic myocardium were also measured and compared. RESULTS: The construction of MI model resulted in a LVEF reduction of 50% compared with the sham-control. After 28 days, the LVEF value was 10% higher when calycosin (4 mg/kg) was administered compared with the DMSO group. The expression of VEGF and CD31 showed a dose-dependent manner when calycosin was administrated. The calycosin-treated (4 mg/kg) group displayed a two-fold increase in VEGF expression at both the mRNA and protein levels compared with the DMSO group. In addition, CD31 expression in the microvascular increased 1.5-fold in the 4 mg/kg calycosin-treated group. CONCLUSION: Calycosin improved left ventricular ejection fraction in the MI rat models, induced VEGF expression in the ischaemic myocardium, increased CD31 expression and promoted angiogenesis.


Asunto(s)
Medicamentos Herbarios Chinos/administración & dosificación , Ventrículos Cardíacos/fisiopatología , Isoflavonas/administración & dosificación , Infarto del Miocardio/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Humanos , Masculino , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Ratas , Ratas Sprague-Dawley , Volumen Sistólico/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
IEEE Trans Biomed Eng ; 62(1): 4-20, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25389236

RESUMEN

The use of irreversible electroporation (IRE) for cancer treatment has increased sharply over the past decade. As a nonthermal therapy, IRE offers several potential benefits over other focal therapies, which include 1) short treatment delivery time, 2) reduced collateral thermal injury, and 3) the ability to treat tumors adjacent to major blood vessels. These advantages have stimulated widespread interest in basic through clinical studies of IRE. For instance, many in vitro and in vivo studies now identify treatment planning protocols (IRE threshold, pulse parameters, etc.), electrode delivery (electrode design, placement, intraoperative imaging methods, etc.), injury evaluation (methods and timing), and treatment efficacy in different cancer models. Therefore, this study reviews the in vitro, translational, and clinical studies of IRE cancer therapy based on major experimental studies particularly within the past decade. Further, this study provides organized data and facts to assist further research, optimization, and clinical applications of IRE.


Asunto(s)
Permeabilidad de la Membrana Celular/efectos de la radiación , Electroquimioterapia/métodos , Neoplasias/fisiopatología , Neoplasias/terapia , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Animales , Terapia por Estimulación Eléctrica/métodos , Campos Electromagnéticos , Medicina Basada en la Evidencia , Humanos , Modelos Biológicos , Neoplasias/patología , Neovascularización Patológica/fisiopatología , Resultado del Tratamiento
16.
J Theor Biol ; 355: 194-207, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-24751927

RESUMEN

Vascularized tumor growth is characterized by both abnormal interstitial fluid flow and the associated interstitial fluid pressure (IFP). Here, we study the effect that these conditions have on the transport of therapeutic agents during chemotherapy. We apply our recently developed vascular tumor growth model which couples a continuous growth component with a discrete angiogenesis model to show that hypertensive IFP is a physical barrier that may hinder vascular extravasation of agents through transvascular fluid flux convection, which drives the agents away from the tumor. This result is consistent with previous work using simpler models without blood flow or lymphatic drainage. We consider the vascular/interstitial/lymphatic fluid dynamics to show that tumors with larger lymphatic resistance increase the agent concentration more rapidly while also experiencing faster washout. In contrast, tumors with smaller lymphatic resistance accumulate less agents but are able to retain them for a longer time. The agent availability (area-under-the curve, or AUC) increases for less permeable agents as lymphatic resistance increases, and correspondingly decreases for more permeable agents. We also investigate the effect of vascular pathologies on agent transport. We show that elevated vascular hydraulic conductivity contributes to the highest AUC when the agent is less permeable, but to lower AUC when the agent is more permeable. We find that elevated interstitial hydraulic conductivity contributes to low AUC in general regardless of the transvascular agent transport capability. We also couple the agent transport with the tumor dynamics to simulate chemotherapy with the same vascularized tumor under different vascular pathologies. We show that tumors with an elevated interstitial hydraulic conductivity alone require the strongest dosage to shrink. We further show that tumors with elevated vascular hydraulic conductivity are more hypoxic during therapy and that the response slows down as the tumor shrinks due to the heterogeneity and low concentration of agents in the tumor interior compared with the cases where other pathological effects may combine to flatten the IFP and thus reduce the heterogeneity. We conclude that dual normalizations of the micronevironment - both the vasculature and the interstitium - are needed to maximize the effects of chemotherapy, while normalization of only one of these may be insufficient to overcome the physical resistance and may thus lead to sub-optimal outcomes.


Asunto(s)
Antineoplásicos , Presión Sanguínea , Linfangiogénesis , Modelos Biológicos , Modelos Teóricos , Neoplasias , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Transporte Biológico Activo , Humanos , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/psicología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología
17.
ScientificWorldJournal ; 2014: 919570, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24563633

RESUMEN

Blood supply is essential for development and growth of tumors and angiogenesis is the fundamental process of new blood vessel formation from preexisting ones. Angiogenesis is a prognostic indicator for a variety of tumors, and it coincides with increased shedding of neoplastic cells into the circulation and metastasis. Several molecules such as cell surface receptors, growth factors, and enzymes are involved in this process. While antiangiogenic therapy for cancer has been proposed over 20 years ago, it has garnered much controversy in recent years within the scientific community. The complex relationships between the angiogenic signaling cascade and antiangiogenic substances have indicated the angiogenic pathway as a valid target for anticancer drug development and VEGF has become the primary antiangiogenic drug target. This review discusses the basic and clinical perspectives of angiogenesis highlighting the importance of comparative biology in understanding tumor angiogenesis and the integration of these model systems for future drug development.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Humanos , Neoplasias/fisiopatología , Células Madre Neoplásicas/metabolismo , Neovascularización Patológica/fisiopatología , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
PLoS One ; 9(1): e85891, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24465768

RESUMEN

Local renin-angiotensin systems exist in various malignant tumor tissues; this suggests that the main effector peptide, angiotensin II, could act as a key factor in tumor growth. The underlying mechanisms for the anti-angiogenic effect of angiotensin II type 1 receptor blockers need to be further evaluated. The present study was carried out to investigate the anti-angiogenic effect of olmesartan alone or in combination with sorafenib, an angiotensin (1-7) agonist or an angiotensin (1-7) antagonist in Ehrlich's ascites carcinoma-bearing mice. The tumor was induced by intradermal injection of Ehrlich's ascites carcinoma cells into mice. Tumor discs were used to evaluate the microvessel density; the serum levels of vascular endothelial growth factor (VEGF) and serum insulin-like growth factor I (IGF-I); and their intratumoral receptors, VEGF receptor-2 and IGF-I receptor, respectively. All parameters were determined following the treatment course, which lasted for 21 days post-inoculation. Monotherapy with olmesartan and its combination with sorafenib resulted in a significant reduction in microvessel density and serum levels of VEGF and IGF-I, as well as their intratumoral receptors. In addition, the combination of olmesartan (30 mg/kg) with an angiotensin (1-7) agonist reduced the microvessel density, IGF-I serum levels and the levels of its intratumoral receptor. In conclusion, olmesartan reduced the levels of the angiogenesis markers IGF-I and VEGF and down-regulated the intratumoral expression of their receptors in a dose-dependent manner, and these effects were dependent on the angiotensin (1-7) receptor. These results suggest that olmesartan is a promising adjuvant to sorafenib in the treatment of cancer.


Asunto(s)
Carcinoma de Ehrlich/tratamiento farmacológico , Imidazoles/farmacología , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Tetrazoles/farmacología , Angiotensina I/agonistas , Angiotensina I/antagonistas & inhibidores , Angiotensina I/fisiología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Antineoplásicos/farmacología , Carcinoma de Ehrlich/metabolismo , Carcinoma de Ehrlich/fisiopatología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Inmunohistoquímica , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Neovascularización Patológica/prevención & control , Niacinamida/farmacología , Fragmentos de Péptidos/agonistas , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/fisiología , Receptor IGF Tipo 1/metabolismo , Sorafenib , Carga Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/sangre , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
19.
BMC Complement Altern Med ; 13: 168, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23842450

RESUMEN

BACKGROUND: Syzygium campanulatum Korth (Myrtaceae) is an evergreen shrub rich in phenolics, flavonoid antioxidants, and betulinic acid. This study sought to investigate antiangiogenic and anti-colon cancer effects of S.C. standardized methanolic extract. METHODS: Betulinic acid was isolated from methanolic extract by crystallization and chromatography techniques. S.C. methanolic extract was analyzed by UV-Vis spectrophotometry, FTIR, LC-MS, and HPLC. Antiangiogenic effect was studied on rat aortic rings, matrigel tube formation, cell proliferation and migration, and expression of vascular endothelial growth factor (VEGF). Antitumor effect was studied using a subcutaneous tumor model of HCT 116 colorectal carcinoma cells established in nude mice. RESULTS: Analysis by HPLC, LC-MS and FTIR confirm presence of betulinic acid in S.C. methanolic extract. Quantitative analysis by HPLC indicates presence of betulinic acid in S.C. extract at 5.42 ± 0.09% (w/w). Antiangiogenesis study showed potent inhibition of microvessels outgrowth in rat aortic rings, and studies on normal and cancer cells did not show any significant cytotoxic effect. Antiangiogenic effect was further confirmed by inhibition of tube formation on matrigel matrix that involves human endothelial cells (IC50 = 17.6 ± 2.9 µg/ml). S.C. extract also inhibited migration of endothelial cells and suppressed expression of VEGF. In vivo antiangiogenic study showed inhibition of new blood vessels in chicken embryo chorioallantoic membrane (CAM), and in vivo antitumor study showed significant inhibition of tumor growth due to reduction of intratumor blood vessels and induction of cell death. CONCLUSION: Collectively, our results indicate S. campanulatum as antiangiogenic and antitumor candidate, and a new source of betulinic acid.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Regulación hacia Abajo/efectos de los fármacos , Inhibidores de Crecimiento/administración & dosificación , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Syzygium/química , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/aislamiento & purificación , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Femenino , Inhibidores de Crecimiento/química , Inhibidores de Crecimiento/aislamiento & purificación , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/fisiopatología , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Ratas , Ratas Sprague-Dawley , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Circ Res ; 113(4): 408-17, 2013 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-23770613

RESUMEN

RATIONALE: Growing evidence indicates that oxidative stress contributes markedly to endothelial dysfunction. The selenoenzyme glutathione peroxidase 4 (Gpx4) is an intracellular antioxidant enzyme important for the protection of membranes by its unique activity to reduce complex hydroperoxides in membrane bilayers and lipoprotein particles. Yet a role of Gpx4 in endothelial cell function has remained enigmatic. OBJECTIVE: To investigate the role of Gpx4 ablation and subsequent lipid peroxidation in the vascular compartment in vivo. METHODS AND RESULTS: Endothelium-specific deletion of Gpx4 had no obvious impact on normal vascular homeostasis, nor did it impair tumor-derived angiogenesis in mice maintained on a normal diet. In stark contrast, aortic explants from endothelium-specific Gpx4 knockout mice showed a markedly reduced number of endothelial branches in sprouting assays. To shed light onto this apparent discrepancy between the in vivo and ex vivo results, we depleted mice of a second antioxidant, vitamin E, which is normally absent under ex vivo conditions. Therefore, mice were fed a vitamin E-depleted diet for 6 weeks before endothelial deletion of Gpx4 was induced by 4-hydroxytamoxifen. Surprisingly, ≈80% of the knockout mice died. Histopathological analysis revealed detachment of endothelial cells from the basement membrane and endothelial cell death in multiple organs, which triggered thrombus formation. Thromboembolic events were the likely cause of various clinical pathologies, including heart failure, renal and splenic microinfarctions, and paraplegia. CONCLUSIONS: Here, we show for the first time that in the absence of Gpx4, sufficient vitamin E supplementation is crucial for endothelial viability.


Asunto(s)
Glutatión Peroxidasa/deficiencia , Glutatión Peroxidasa/genética , Trombosis/etiología , Trombosis/mortalidad , Deficiencia de Vitamina E/complicaciones , Vitamina E/genética , Animales , Apoptosis/fisiología , Presión Sanguínea/fisiología , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Femenino , Glutatión Peroxidasa/metabolismo , Frecuencia Cardíaca/fisiología , Peroxidación de Lípido/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neovascularización Patológica/fisiopatología , Estrés Oxidativo/fisiología , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Trombosis/fisiopatología , Vitamina E/metabolismo , Deficiencia de Vitamina E/metabolismo , Deficiencia de Vitamina E/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA