Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLOS Digit Health ; 1(11): e0000036, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36812590

RESUMO

Here we describe the design and implementation of a novel data management platform for an academic cancer center which meets the needs of multiple stakeholders. A small, cross-functional technical team identified key challenges to creating a broad data management and access software solution: lowering the technical skill floor, reducing cost, enhancing user autonomy, optimizing data governance, and reimagining technical team structures in academia. The Hyperion data management platform was designed to meet these challenges in addition to usual considerations of data quality, security, access, stability, and scalability. Implemented between May 2019 and December 2020 at the Wilmot Cancer Institute, Hyperion includes a sophisticated custom validation and interface engine to process data from multiple sources, storing it in a database. Graphical user interfaces and custom wizards permit users to directly interact with data across operational, clinical, research, and administrative contexts. The use of multi-threaded processing, open-source programming languages, and automated system tasks (normally requiring technical expertise) minimizes costs. An integrated ticketing system and active stakeholder committee support data governance and project management. A co-directed, cross-functional team with flattened hierarchy and integration of industry software management practices enhances problem solving and responsiveness to user needs. Access to validated, organized, and current data is critical to the functioning of multiple domains in medicine. Although there are downsides to developing in-house customized software, we describe a successful implementation of custom data management software in an academic cancer center.

2.
Mol Med ; 16(1-2): 10-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19946606

RESUMO

The main circulating estrogen hormone 17beta-estradiol (E2) contributes to the initiation and progression of breast cancer. Estrogen receptors (ERs), as transcription factors, mediate the effects of E2. Ablation of the circulating E2 and/or prevention of ER functions constitute approaches for ER-positive breast cancer treatments. These modalities are, however, ineffective in de novo endocrine-resistant breast neoplasms that do not express ERs. The interaction of E2-ERs with specific DNA sequences, estrogen responsive elements (EREs), of genes constitutes one genomic pathway necessary for cellular alterations. We herein tested the prediction that specific regulation of ERE-driven genes by an engineered monomeric and constitutively active transcription factor, monotransregulator, provides a basis for the treatment of ER-negative breast cancer. Using adenovirus infected ER-negative MDA-MB-231 cells derived from a breast adenocarcinoma, we found that the monotransregulator, but not the ERE-binding defective counterpart, repressed cellular proliferation and motility, and induced apoptosis through expression of genes that required ERE interactions. Similarly, the monotransregulator suppressed the growth of ER-negative BT-549 cells derived from a breast-ductal carcinoma. Moreover, the ERE-binding monotransregulator repressed xenograft tumor growth in a nude mice model. Thus, specific regulation of genes bearing EREs could offer a therapeutic approach for de novo endocrine-resistant breast cancers.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/terapia , Receptor alfa de Estrogênio/genética , Elementos de Resposta , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Morte Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Estradiol/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Genes Reguladores , Terapia Genética/métodos , Humanos , Camundongos , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Radiat Res ; 171(5): 606-14, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19580496

RESUMO

A variety of antiangiogenic strategies have proven effective in preclinical tumor models, either as single agents or in combination with radiation. Clinical gains have been relatively modest, however, and questions remain regarding optimal scheduling. The objectives of the current work were to evaluate whether the sequencing of acute treatment critically affects tumor pathophysiological and therapeutic response. Axitinib (Pfizer Global Research & Development), an inhibitor that predominantly targets vascular endothelial growth factor receptors, was administered either before or after each daily radiation fraction in two human prostate xenograft tumor models. Tumors were frozen at sequential times to monitor changes in (1) vascular spacing, (2) pericyte and basement membrane coverage, and (3) hypoxia. Although similar reductions in blood vessel counts were observed with each tumor model, tumor vasculature was not functionally normalized. Instead, tumor hypoxia increased, accompanied by a progressive dissociation of pericytes and basement membranes. Ultimately, tumor growth inhibition was found to be equivalent for each of the combination schedules. These studies illustrate a clear advantage to combining axitinib with fractionated therapy but argue against an acute radiosensitization or radioprotection of either the tumor cells or tumor vasculature. Instead, post- and preirradiation daily drug administration serve equally well in supplementing the response to radiotherapy.


Assuntos
Fracionamento da Dose de Radiação , Imidazóis/farmacologia , Indazóis/farmacologia , Neoplasias/terapia , Inibidores de Proteínas Quinases/farmacologia , Axitinibe , Membrana Basal/efeitos dos fármacos , Membrana Basal/efeitos da radiação , Linhagem Celular Tumoral , Terapia Combinada , Humanos , Masculino , Neoplasias/patologia , Pericitos/efeitos dos fármacos , Pericitos/efeitos da radiação
4.
Cancer Res ; 67(20): 9921-8, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17942924

RESUMO

Although antiangiogenic strategies have proven highly promising in preclinical studies and some recent clinical trials, generally only combinations with cytotoxic therapies have shown clinical effectiveness. An ongoing question has been whether conventional therapies are enhanced or compromised by antiangiogenic agents. The present studies were designed to determine the pathophysiologic consequences of both single and combined treatments using fractionated radiotherapy plus AG-013736, a receptor tyrosine kinase inhibitor that preferentially inhibits vascular endothelial growth factor receptors. DU145 human prostate xenograft tumors were treated with (a) vehicle alone, (b) AG-013736, (c) 5x2 Gy/wk radiotherapy fractions, or (d) the combination. Automated image processing of immunohistochemical images was used to determine total and perfused blood vessel spacing, overall hypoxia, pericyte/collagen coverage, proliferation, and apoptosis. Combination therapy produced an increased tumor response compared with either monotherapy alone. Vascular density progressively declined in concert with slightly increased alpha-smooth muscle actin-positive pericyte coverage and increased overall tumor hypoxia (compared with controls). Although functional vessel endothelial apoptosis was selectively increased, reductions in total and perfused vessels were generally proportionate, suggesting that functional vasculature was not specifically targeted by combination therapy. These results argue against either an AG-013736- or a combination treatment-induced functional normalization of the tumor vasculature. Vascular ablation was mirrored by the increased appearance of dissociated pericytes and empty type IV collagen sleeves. Despite the progressive decrease in tumor oxygenation over 3 weeks of treatment, combination therapy remained effective and tumor progression was minimal.


Assuntos
Imidazóis/farmacologia , Indazóis/farmacologia , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/terapia , Actinas/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Axitinibe , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos da radiação , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/efeitos da radiação , Colágeno Tipo IV/metabolismo , Terapia Combinada , Fracionamento da Dose de Radiação , Humanos , Masculino , Camundongos , Camundongos Nus , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Neovascularização Patológica/radioterapia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Clin Cancer Res ; 13(16): 4891-9, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17699869

RESUMO

PURPOSE: The lack of effective treatment for pancreatic cancer results in a very low survival rate. This study explores the enhancement of the therapeutic effect on human pancreatic cancer via the combination of triptolide and ionizing radiation (IR). EXPERIMENTAL DESIGN: In vitro AsPC-1 human pancreatic cancer cells were treated with triptolide alone, IR alone, or triptolide plus IR. Cell proliferation was analyzed with sulforhodamine B (SRB) method and clonogenic survival; comparison of apoptosis induced by the above treatment was analyzed by annexin V-propidium iodide (PI) staining. Furthermore, the expression of apoptotic pathway intermediates was measured by the assay of caspase activity and Western blot. Mitochondrial transmembrane potential was determined by JC-1 assay. In vivo, AsPC-1 xenografts were treated with 0.25 mg/kg triptolide, 10 Gy IR, or triptolide plus IR. The tumors were measured for volume and weight at the end of the experiment. Tumor tissues were tested for terminal nucleotidyl transferase-mediated nick end labeling (TUNEL) and immunohistochemistry. RESULTS: The combination of triptolide plus IR reduced cell survival to 21% and enhanced apoptosis, compared with single treatment. In vivo, tumor growth of AsPC-1 xenografts was reduced further in the group treated with triptolide plus IR compared with single treatment. TUNEL and immunohistochemistry of caspase-3 cleavage in tumor tissues indicated that the combination of triptolide plus IR resulted in significantly enhanced apoptosis compared with single treatments. CONCLUSIONS: Triptolide in combination with ionizing radiation produced synergistic antitumor effects on pancreatic cancer both in vitro and in vivo and seems promising in the combined modality therapy of pancreatic cancer.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Diterpenos/farmacologia , Neoplasias Pancreáticas/terapia , Fenantrenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Terapia Combinada , Compostos de Epóxi/farmacologia , Fase G2/efeitos dos fármacos , Fase G2/efeitos da radiação , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Pancreáticas/radioterapia , Transplante Heterólogo
6.
Adv Exp Med Biol ; 566: 59-65, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16594135

RESUMO

Since conventional therapies are directly dependent on the supply of either drugs or oxygen, a key question is whether antiangiogenic agents produce detrimental effects on tumor vascular function, thus compromising combination therapies. A second question is whether experimental results based on fast-growing, transplanted tumors mimic those in slowly developing spontaneous tumors, which may be more representative of response in human primary tumors. To investigate changes in tumor pathophysiology, three antiangiogenic agents were compared: a) endostatin, b) anti-VEGFR-2 (DC101), and c) celecoxib. Total blood vessels were identified using anti-CD31, perfused vessels using DiOC7, and hypoxia by EF5 uptake. Although individual tumor growth rates varied substantially, DC101 produced the most striking inhibition. DC101 increased total and perfused vessel spacing as well as overall hypoxia, while endostatin increased total vessel spacing, and hypoxia and celecoxib had no marked effects. These results reinforce the idea that pathophysiological changes in spontaneous tumors are in general reflective of response in transplanted tumors. Furthermore, although DC101 inhibited growth in roughly half of the spontaneous tumors, the remaining tumors were unaffected. A key focus of future studies will be to investigate the underlying rationale for the widely varying antiangiogenic response among tumors that outwardly appear so similar.


Assuntos
Inibidores da Angiogênese/farmacologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Animais , Celecoxib , Endostatinas/farmacologia , Feminino , Hipóxia/tratamento farmacológico , Hipóxia/fisiopatologia , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/fisiopatologia , Camundongos , Camundongos Endogâmicos C3H , Pirazóis/farmacologia , Proteínas Recombinantes/farmacologia , Sulfonamidas/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
7.
Cancer Res ; 64(16): 5712-9, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15313911

RESUMO

Although clinical trials of antiangiogenic strategies have been disappointing when administered as single agents, such approaches can play an important role in cancer treatment when combined with conventional therapies. Previous studies have shown that DC101, an antiangiogenic monoclonal antibody against vascular endothelial growth factor receptor-2, can produce significant growth inhibition in spontaneous and transplanted tumors but can also induce substantial hypoxia. Because DC101 appears to potentiate radiotherapy in some tumors, the present studies were undertaken to characterize pathophysiological changes following combined therapy and to determine whether radioresponse is enhanced despite the induction of hypoxia. MCa-4 and MCa-35 mammary carcinomas were treated with: (a) DC101; (b) 5 x 6 Gy radiation fractions; or (c) the combination. Image analysis of frozen tumor sections was used to quantitate: (a) hypoxia; (b) spacing of total and perfused blood vessels; and (c) endothelial and tumor cell apoptosis. For MCa-4, combination treatment schedules produced significant and prolonged delays in tumor growth, whereas single-modality treatments had minor effects. For MCa-35, radiation or the combination led to equivalent growth inhibition. In all tumors, hypoxia increased markedly after either radiation or DC101 alone. Although combination therapy produced no immediate pathophysiological changes, hypoxia ultimately increased after cessation of therapy. Preferential increases in endothelial apoptosis following combination treatment suggest that in addition to blocking tumor angiogenesis, DC101 enhances radiotherapy by specifically sensitizing endothelial cells, leading to degeneration of newly formed blood vessels.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias Mamárias Experimentais/radioterapia , Neoplasias Mamárias Experimentais/terapia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Divisão Celular/fisiologia , Divisão Celular/efeitos da radiação , Hipóxia Celular/fisiologia , Hipóxia Celular/efeitos da radiação , Terapia Combinada , Fracionamento da Dose de Radiação , Endotélio Vascular/patologia , Endotélio Vascular/efeitos da radiação , Feminino , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos C3H , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia
8.
Radiother Oncol ; 72(2): 221-30, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15297140

RESUMO

BACKGROUND AND PURPOSE: The primary objectives of this study were to address two major questions. (1) Does VEGF receptor-2 antibody (DC101) produce detrimental effects on tumor vascular function and oxygenation that could compromise adjuvant therapies? (2) Is pathophysiological response to such antiangiogenic strategies different in transplanted versus primary spontaneous tumors? MATERIALS AND METHODS: The effects of early and late initiation DC101 treatment were evaluated using spontaneous murine mammary carcinomas and two markedly different transplanted mammary tumors, MCa-35 and MCa-4. Mice were administered DC101 or saline, tumors were frozen, and immunohistochemical staining was quantified using image analysis of multiply-stained frozen sections. Total blood vessels were identified using antibodies to CD31 or panendothelial antigen, perfused vessels via i.v. injection of fluorescent DiOC7, and tumor hypoxia by hypoxia marker (EF5) uptake. RESULTS: Tumor growth was significantly inhibited following DC101 administration in all tumor models. In general, early initiation DC101 treatment reduced perfused vessel counts and increased tumor hypoxia, while late initiation treatment had no significant impact on either. Results indicate that DC101 slows tumor growth through a decrease in vascular function, leading to increased tumor cell apoptosis and necrosis at sites distant from perfused blood vessels, and suggest that DC101 accelerates the rate at which tumor cells outgrow their functional vascular supply. CONCLUSIONS: Although highly variable among individual spontaneous tumors, the overall effects of DC101 on tumor hypoxia were quite similar between spontaneous and transplanted tumors. Since reductions in tumor oxygenation due to antiangiogenic treatment were transient, initial pathophysiological deficiencies that could compromise conventional therapies over the short-term may be of less relevance when administered over more extended treatment schedules.


Assuntos
Neoplasias da Mama/imunologia , Hipóxia Celular/imunologia , Endotélio Vascular/imunologia , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia , Animais , Anticorpos/administração & dosagem , Linhagem Celular Tumoral , Feminino , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos C3H , Transplante de Neoplasias
9.
Adv Exp Med Biol ; 530: 165-76, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14562714

RESUMO

Since quantitative measurements of tumor vascular function cannot be obtained in human tumors, appropriate animal tumor models must be utilized. The current studies were undertaken to compare transplantable, murine KHT tumors with primary and 1st generation transplants of spontaneous mammary carcinomas. To evaluate changes in tumor vascular structure and function, immunostaining of total and perfused vascular spacing, and cryospectrophotometric measurement of intravascular HbO2 saturations were utilized. KHT tumors demonstrated a distinct pattern of decreasing oxygenation with increasing distance from the tumor surface, while spontaneous tumors exhibited striking intertumor heterogeneities and a reduced dependence of oxygenation on distance from tumor surface. Anatomical/perfused vessel distributions and functional response were similar between the primary and transplanted tumor models, as was tissue histological appearance, but were quite different from KHT tumors. These results indicate that spontaneous tumor vascular configuration and function tend to be preserved in 1st generation trochar transplanted tumors.


Assuntos
Neoplasias Mamárias Experimentais/metabolismo , Oxigênio/metabolismo , Animais , Imuno-Histoquímica , Neoplasias Mamárias Experimentais/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C3H , Transplante de Neoplasias , Fluxo Sanguíneo Regional
10.
Adv Exp Med Biol ; 530: 603-9, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14562757

RESUMO

Isotransplants of murine fibrosarcoma (KHT) cells were inoculated i.m. into the hind limbs of 6-8 week-old female C3H/HeJ mice. Intratumoral injection of FGF1 or VEGF proteins decreased hypoxic marker uptake in murine fibrosarcoma KHT. Reduction of tumor hypoxia did not correlate with mRNA expression of transcription factors in tumors. Likewise, there was no significant alteration in either apoptotic frequency or the mRNA levels of 10 apoptotic-related molecules in FGF1- or VEGF-treated tumors. mRNA expression for MCP-1, IL-1 beta, IL-18, and IL-1Ra, however, were decreased in the tumors following FGF1 or VEGF treatment. Among the normal tissues tested (brain, kidney, liver, spleen, and lung), basal mRNA levels for cytokines and chemokines varied. Intratumoral injection of FGF1 or VEGF (6 daily intra-tumor injections of 6 micrograms/mouse) did not alter most cytokine or chemokine mRNA expression in spleen and lung. In summary, alteration of tumor oxygenation by local administration of angiogenic growth factors may be mediated by cytokine/chemokine production in the tumor.


Assuntos
Fator 1 de Crescimento de Fibroblastos/fisiologia , Fibrossarcoma/patologia , Neovascularização Patológica , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Linhagem Celular Tumoral , Feminino , Camundongos , Camundongos Endogâmicos C3H , Transplante de Neoplasias
11.
Int J Radiat Oncol Biol Phys ; 57(4): 1038-46, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14575835

RESUMO

PURPOSE: Recent results in the literature have demonstrated that the antiangiogenic agent endostatin can enhance antitumor effects when administered before or during radiotherapy. To better understand the underlying pathophysiologic basis for this radiosensitization, the current study investigated whether short-term endostatin administration is linked to alterations in tumor vascular perfusion and oxygen delivery. METHODS AND MATERIALS: Three daily doses of recombinant endostatin (20 mg/kg) were administered to two murine mammary carcinomas, the highly vascularized MCa-35 and the less vascularized MCa-4. Image analysis techniques were used to quantify (1) total and perfused vascular spacing, and (2) changes in tumor hypoxia as a function of distance from the nearest blood vessel. RESULTS: In MCa-35 tumors, endostatin had no effect on vessel spacing, tumor hypoxia, or tumor growth. In MCa-4 tumors, total and perfused vessel spacings were also unchanged, but tumor growth was inhibited, and tumor hypoxia significantly decreased. These tumors demonstrated an increased vascular functionality suggestive of an increase in the number of intermittently perfused vessels, without corresponding alterations in tumor oxygen consumption rate. CONCLUSIONS: Poorly vascularized, hypoxic mammary carcinomas were much more responsive to short-term endostatin treatment than well-vascularized, more homogeneously oxygenated tumors. Oxygen levels in the responsive tumors were transiently improved after treatment, which could have substantial implications with respect to the therapeutic effectiveness of combining antiangiogenic agents with conventional therapies.


Assuntos
Inibidores da Angiogênese/farmacologia , Hipóxia Celular/efeitos dos fármacos , Endostatinas/farmacologia , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neovascularização Patológica/tratamento farmacológico , Animais , Apoptose , Carbocianinas , Divisão Celular , Avaliação Pré-Clínica de Medicamentos , Feminino , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos C3H
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...