Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Oncol Pharm Pract ; 28(8): 1763-1770, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34569871

RESUMO

OBJECTIVE: A prospective open-label randomized controlled trial to assess the role of a picture-based medication calendar on adherence to antiemetic regimens for adult patients receiving chemotherapy and assess the effect on other medication taking behaviors as well as patient satisfaction with the tool. METHODS: Participants were randomly assigned 1:1 to routine care with or without calendar. RESULTS: Adherence, stratified by education (university or postgraduate, p = 0.09; grade school, high school or college p = 0.32), was non-significantly different between study arms. At least 70% of intervention arm participants moderately or completely agreed that the calendar helped with medication taking behaviors. There was no statistical difference between study arms for perceived regimen complexity (p = 0.16). Medication Use and Self Efficacy score (adjusted for age) used to assess perceived self-efficacy with medication taking behaviors were not statistically significant between study arms (p = 0.09). CONCLUSION: The picture-based medication calendar did not statistically affect adherence to scheduled antiemetics among outpatients receiving chemotherapy for solid organ tumor origins. However, participants indicated that the calendar was effective for keeping track of medications, had an easy-to-understand layout, and provided help around when and how to take medications related to the oncology regimen.


Assuntos
Antieméticos , Neoplasias , Adulto , Humanos , Antieméticos/uso terapêutico , Estudos Prospectivos , Neoplasias/tratamento farmacológico , Adesão à Medicação
2.
Tumour Biol ; 43(1): 285-306, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34690152

RESUMO

Peripheral human blood is a readily-accessible source of patient material in which circulating tumour cells (CTCs) can be found. Their isolation and characterization holds the potential to provide prognostic value for various solid cancers. Enumeration of CTCs from blood is becoming a common practice in informing prognosis and may guide therapy decisions. It is further recognized that enumeration alone does not capture perspective on the heterogeneity of tumours and varying functional abilities of the CTCs to interact with the secondary microenvironment. Characterizing the isolated CTCs further, in particular assessing their functional abilities, can track molecular changes in the disease progress. As a step towards identifying a suite of functional features of CTCs that could aid in clinical decisions, developing a CTC isolation technique based on extracellular matrix (ECM) interactions may provide a more solid foundation for isolating the cells of interest. Techniques based on size, charge, density, and single biomarkers are not sufficient as they underutilize other characteristics of cancer cells. The ability of cancer cells to interact with ECM proteins presents an opportunity to utilize their full character in capturing, and also allows assessment of the features that reveal how cells might behave at secondary sites during metastasis. This article will review some common techniques and recent advances in CTC capture technologies. It will further explore the heterogeneity of the CTC population, challenges they experience in their metastatic journey, and the advantages of utilizing an ECM-based platform for CTC capture. Lastly, we will discuss how tailored ECM approaches may present an optimal platform to capture an influential heterogeneous population of CTCs.


Assuntos
Metástase Neoplásica/patologia , Células Neoplásicas Circulantes/patologia , Biomarcadores Tumorais/metabolismo , Carcinoma/secundário , Adesão Celular , Separação Celular , Transição Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Humanos , Metástase Neoplásica/diagnóstico , Células Neoplásicas Circulantes/metabolismo , Prognóstico
3.
Pharm Res ; 34(12): 2579-2595, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28924691

RESUMO

Antibody-drug conjugates (ADCs) are ushering in the next era of targeted therapy against cancer. An ADC for cancer therapy consists of a potent cytotoxic payload that is attached to a tumour-targeted antibody by a chemical linker, usually with an average drug-to-antibody ratio (DAR) of 3.5-4. The theory is to deliver potent cytotoxic payloads directly to tumour cells while sparing healthy cells. However, practical application has proven to be more difficult. At present there are only two ADCs approved for clinical use. Nevertheless, in the last decade there has been an explosion of options for ADC engineering to optimize target selection, Fc receptor interactions, linker, payload and more. Evaluation of these strategies requires an understanding of the mechanistic underpinnings of ADC pharmacokinetics. Development of ADCs for use in cancer further requires an understanding of tumour properties and kinetics within the tumour environment, and how the presence of cancer as a disease will impact distribution and elimination. Key pharmacokinetic considerations for the successful design and clinical application of ADCs in oncology are explored in this review, with a focus on the mechanistic determinants of distribution and elimination.


Assuntos
Antineoplásicos/farmacocinética , Imunoconjugados/farmacocinética , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Sistemas de Liberação de Medicamentos/métodos , Humanos , Imunoconjugados/administração & dosagem , Imunoconjugados/química , Distribuição Tecidual
4.
BMC Cancer ; 15: 882, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26552750

RESUMO

BACKGROUND: Recurrence of colorectal cancer (CRC) may arise due to the persistence of drug-resistant and cancer-initiating cells that survive exposure to chemotherapy. Proteins responsible for this recurrence include the chemokine receptor CXCR4, which is known to enable CRC metastasis, as well as the cancer-initiating cell marker and peptidase CD26, which terminates activity of its chemokine CXCL12. METHODS: We evaluated the expression and function of CXCR4 and CD26 in colon cancer cell lines and xenografts following treatment with common chemotherapies using radioligand binding, flow cytometry, immunofluorescence, and enzymatic assays. RESULTS: 5-Fluorouracil, oxaliplatin and SN-38 (the active metabolite of irinotecan), as well as cisplatin, methotrexate and vinblastine, each caused decreases in cell-surface CXCR4 and concomitant increases in CD26 on HT-29, T84, HRT-18, SW480 and SW620 CRC cell lines. Flow cytometry indicated that the decline in CXCR4 was associated with a significant loss of CXCR4+/CD26- cells. Elevations in CD26 were paralleled by increases in both the intrinsic dipeptidyl peptidase activity of CD26 as well as its capacity to bind extracellular adenosine deaminase. Orthotopic HT-29 xenografts treated with standard CRC chemotherapeutics 5-fluorouracil, irinotecan, or oxaliplatin showed dramatic increases in CD26 compared to untreated tumors. Consistent with the loss of CXCR4 and gain in CD26, migratory responses to exogenous CXCL12 were eliminated in cells pretreated with cytotoxic agents, although cells retained basal motility. Analysis of cancer-initiating cell CD44 and CD133 subsets revealed drug-dependent responses of CD26/CD44/CD133 populations, suggesting that the benefits of combining standard chemotherapies 5-fluoruracil and oxaliplatin may be derived from their complementary elimination of cell populations. CONCLUSION: Our results indicate that conventional anticancer agents may act to inhibit chemokine-mediated migration through eradication of CXCR4+ cells and attenuation of chemokine gradients through elevation of CD26 activity.


Assuntos
Quimiocina CXCL12/biossíntese , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Dipeptidil Peptidase 4/biossíntese , Receptores CXCR4/biossíntese , Animais , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Carcinogênese/efeitos dos fármacos , Linhagem da Célula , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL12/genética , Neoplasias do Colo/patologia , Dipeptidil Peptidase 4/genética , Fluoruracila/administração & dosagem , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HT29 , Humanos , Receptores de Hialuronatos/genética , Irinotecano , Camundongos , Metástase Neoplásica , Receptores CXCR4/genética , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Cancer Rep (Hoboken) ; 5(2): e1475, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34313032

RESUMO

BACKGROUND: The morbidity and mortality of cancer are significantly impacted by the invasive and metastatic potential of particular subgroups of malignant cells within a tumor. The particular pre-metastatic properties of cancerous cells are thus a critical target for novel therapeutics in the oncology field. Cannabinoid molecules have been investigated in recent years in the context of invasion and metastasis of various malignancies, with varying effects reported in the literature. RECENT FINDINGS: There was substantial variability in the findings reported by the literature of the effects of cannabinoid molecules on cancer cell invasion and metastasis. These effects varied depending on which ligand and which of the CB1, CB2, or GPR55 receptors were investigated. These findings suggest a role for the phenomenon of biased signaling in explaining the diversity of effects of cannabinoid molecules on cancer cell invasion. CONCLUSION: While substantially more investigation is required into the effects of cannabinoid molecules on cancer cell invasion and metastasis, we describe in this review the significant diversity in the responses of cancer cells to cannabinoid molecules in terms of their invasive and metastatic capacities.


Assuntos
Canabinoides/farmacologia , Neoplasias/tratamento farmacológico , Transição Epitelial-Mesenquimal , Humanos , Invasividade Neoplásica/prevenção & controle , Metástase Neoplásica/prevenção & controle , Fenótipo , Receptores de Canabinoides , Transdução de Sinais
6.
Front Immunol ; 13: 957233, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36591314

RESUMO

Introduction: Colorectal cancer and other adult solid cancers pose a significant challenge for successful treatment because the tumor microenvironment both hinders the action of conventional therapeutics and suppresses the immune activities of infiltrating leukocytes. The immune suppression is largely the effect of enhanced local mediators such as purine nucleosides and eicosanoids. Genetic approaches have the promise of interfering with these mechanisms of local immunosuppression to allow both intrinsic and therapeutic immunological anticancer processes. Bacterial phages offer a novel means of enabling access into tissues for therapeutic genetic manipulations. Methods: We generated spheroids of fibroblastic and CRC cancer cells to model the 3-dimensional stromal and parenchymal components of colorectal tumours. We used these to examine the access and effects of both wildtype (WT) and epidermal growth factor (EGF)-presenting bacteriophage λ (WT- λ and EGF-λ) as a means of delivery of targeted genetic interventions in solid cancers. We used both confocal microscopy of spheroids exposed to AF488-tagged phages, and the recovery of viable phages as measured by plaque-forming assays to evaluate access; and measures of mitochondrial enzyme activity and cellular ATP to evaluate the outcome on the constituent cells. Results: Using flourescence-tagged derivatives of these bacteriophages (AF488-WT-λ and AF488-EGF-λ) we showed that phage entry into these tumour microenvironments was possible and that the EGF ligand enabled efficient and persistent uptake into the cancer cell mass. EGF-λ became localized in the intracellular portion of cancer cells and was subjected to subsequent cellular processing. The targeted λ phage had no independent effect upon mature tumour spheroids, but interfered with the early formation and growth of cancer tissues without the need for addition of a toxic payload, suggesting that it might have beneficial effects by itself in addition to any genetic intervention delivered to the tumour. Interference with spheroid formation persisted over the duration of culture. Discussion: We conclude that targeted phage technology is a feasible strategy to facilitate delivery into colorectal cancer tumour tissue (and by extension other solid carcinomas) and provides an appropriate delivery vehicle for a gene therapeutic that can reduce local immunosuppression and/or deliver an additional direct anticancer activity.


Assuntos
Bacteriófago lambda , Carcinogênese , Neoplasias Colorretais , Microambiente Tumoral , Humanos , Bacteriófago lambda/genética , Bacteriófago lambda/imunologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/imunologia , Receptores ErbB/genética , Receptores ErbB/imunologia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Carcinogênese/genética , Carcinogênese/imunologia
7.
Front Pharmacol ; 13: 1086894, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36618939

RESUMO

Introduction: CD26/dipeptidyl peptidase IV (DPP4) is a cell-surface glycoprotein present on most epithelial cells that modulates the local response to external signals. We have previously shown that the dietary flavone apigenin (4',5,7-trihydroxyflavone) upregulates cell-surface CD26/DPP4 on human colorectal carcinoma (CRC) cells and regulates its activities. We observed a unique synergistic interaction with the CRC chemotherapeutic agent irinotecan, which through its metabolite SN38 elevates CD26 at doses that are sub-cytotoxic. As SN38 interacts with topoisomerase 1 (Topo1) we evaluated whether apigenin influences Topo1 activity. Methods: We used a radioimmunoassay to selectively measure CD26 at the cell surface of HT-29 cells following various treatments. Topoisomerase 1 mRNA expression was measured by q-RT-PCR and protein abundance by western blot analysis. Direct inhibition of topoisomerase activity was measured using an assay of DNA supercoil relaxation with recombinant human Topo1. The role of Topo1 in the effect of apigenin was shown both pharmacologically and by siRNA silencing of Topo1. Molecular docking analysis was done with SBD computational software using the CDOCKER algorithm. Results: The interplay between apigenin and irinotecan was not observed when apigenin was combined with other chemotherapeutic drugs including the topoisomerase 2 inhibitors doxorubicin or etoposide. There was no enhancement of irinotecan action if apigenin was replaced with its hydroxylated metabolite luteolin (3',4',5,7-tetrahydroxyflavone) or emodin (6-methyl-1,3,8-trihydroxyanthraquinone), which is an inhibitor of the principal kinase target of apigenin, casein kinase 2 (CK2). Apigenin did not alter Topo1 mRNA expression, but siRNA knockdown of functional Topo1 eliminated the effect of apigenin and itself increased CD26 levels. Apigenin inhibited Topo1 activity in intact HT-29 cells and showed comparable inhibition of purified recombinant human Topo1 enzyme activity to that of SN-38, the active metabolite of irinotecan. Apigenin fits into the complex of Topo1 with DNA to directly inhibit Topo1 enzyme activity. Discussion: We conclude that apigenin has a unique fit into the Topo1-DNA functional complex that leads to direct inhibition of Topo1 activity, and suggest that this is the basis for the exceptional interaction with the CRC drug irinotecan. A combined action of these two agents may therefore exert a role to limit local signals that facilitate tumour progression.

8.
Front Pharmacol ; 12: 681477, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34084146

RESUMO

Apigenin (4', 5, 7-trihydroxyflavone) is a plant flavone that has been found to have various actions against cancer cells. We evaluated available evidence to determine whether it is feasible for apigenin to have such effects in human patients. Apigenin taken orally is systemically absorbed and recirculated by enterohepatic and local intestinal pathways. Its bioavailability is in the region of 30%. Once absorbed from the oral route it reaches maximal circulating concentration (Cmax) after a time (Tmax) of 0.5-2.5h, with an elimination half-life (T1/2) averaging 2.52 ± 0.56h. Using a circulating concentration for efficacy of 1-5µmol/L as the target, we evaluated data from both human and rodent pharmacokinetic studies to determine if a therapeutic concentration would be feasible. We find that oral intake of dietary materials would require heroic ingestion amounts and is not feasible. However, use of supplements of semi-purified apigenin in capsule form could reach target blood levels using amounts that are within the range currently acceptable for other supplements and medications. Modified formulations or parenteral injection are suitable but may not be necessary. Further work with direct studies of pharmacokinetics and clinical outcomes are necessary to fully evaluate whether apigenin will contribute to a useful clinical strategy, but given emerging evidence that it may interact beneficially with chemotherapeutic drugs, this is worthy of emphasis. In addition, more effective access to intestinal tissues from the oral route raises the possibility that apigenin may be of particular relevance to gastrointestinal disorders including colorectal cancer.

9.
Food Sci Nutr ; 8(10): 5321-5329, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33133535

RESUMO

CD26/DPPIV is a cell surface glycoprotein found on cells of the intestinal epithelium including those of the colon. We have previously shown that the dietary flavone apigenin (4',5,7-trihydroxyflavone) upregulates CD26/DPPIV on colon cells. Flavonoids such as apigenin interfere with the action of multiple cellular protein kinases and have the capacity to modulate the cell exterior and its ability to interface with the local environment through different signaling pathways. We show here that the ability of apigenin to upregulate CD26/DPPIV is exerted through and requires the activity of casein kinase 2 (CK2). Inhibitors of CK2 that are distinct from apigenin (emodin, 6-methyl-1,3,8-trihydroxyanthraquinone; TBB, 4,5,6,7-tetrabromobenzotriazole; and DRB, 5,6-dichlorobenzimidazole 1-ß-D-ribofuranoside) showed a dose-dependent ability to increase CD26/DPPIV and had the same maximal effect when combined with apigenin at submaximal concentrations. Knockdown of CK2 with siRNA abrogated the ability of apigenin to upregulate CD26/DPPIV. Apigenin treatment of cells had no effect on the levels of CK2 protein, consistent with an inhibition of activity of the enzyme. Apigenin's upregulation of CD26/DPPIV in differentiated human colon epithelial cells depends upon inhibition of CK2 activity. This is a key step in enabling apigenin's ability to regulate the functions of intestinal epithelial cells.

10.
Autophagy ; 15(5): 900-907, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30563411

RESUMO

Macroautophagy/autophagy, a pathway by which cellular components are sequestered and degraded in response to homeostatic and cell stress-related signals, is required to preserve hematopoietic stem and progenitor cell function. Loss of chromosomal regions carrying autophagy genes and decreased autophagy gene expression are characteristic of acute myeloid leukemia (AML) cells. Deficiency of autophagy proteins is also linked to an altered AML metabolic profile; altered metabolism has recently emerged as a potential druggable target in AML. Here, we sought to understand the mitochondria-specific changes that occur in leukemia cells after knockdown of BNIP3L/Nix or SQSTM1/p62, which are two autophagy genes involved in mitochondrial clearance and are downregulated in primary AML cells. Mitochondrial function, as measured by changes in endogenous levels of reactive oxygen species (ROS) and mitochondrial membrane potential, was altered in leukemia cells deficient in these autophagy genes. Further, these AML cells were increasingly sensitive to mitochondria-targeting drugs while displaying little change in sensitivity to DNA-targeting agents. These findings suggest that BNIP3L or SQSTM1 may be useful prognostic markers to identify AML patients suitable for mitochondria-targeted therapies. Abbreviations: AML: acute myeloid leukemia; DHE: dihydroethidium; mtDNA: mitochondrial DNA; NAO: 10-N-nonyl acridine orange; PD: population doubling; R123: rhodamine 123; ROS: reactive oxygen species; TRC: transduced scramble controls.


Assuntos
Antineoplásicos/farmacologia , Antioxidantes/farmacologia , Proteínas de Membrana/genética , Mitocôndrias/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , RNA Interferente Pequeno/farmacologia , Proteína Sequestossoma-1/genética , Proteínas Supressoras de Tumor/genética , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Técnicas de Silenciamento de Genes , Células HL-60 , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas de Membrana/antagonistas & inibidores , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Proteína Sequestossoma-1/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/antagonistas & inibidores
11.
Int J Oncol ; 32(3): 527-35, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18292929

RESUMO

The resistance of many human cancers to immune-based therapies, including adoptive immunotherapy and the administration of therapeutic cancer vaccines, has been attributed to tumor-associated immune suppression, due in part to immunosuppressive molecules located within the tumor microenvironment. Adenosine is a purine nucleoside found within the interstitial fluid of solid tumors at concentrations that are able to inhibit cell-mediated immune responses to tumor cells. It is well established that extracellular adenosine inhibits T lymphocyte activation and effector function, including T cell adhesion to tumor cells and cytotoxic activity, by signaling primarily through A2a and A3 adenosine receptors on the surface of T cells. Importantly, A2a adenosine receptor-deficient mice exhibit enhanced anti-tumor immune responses by CD8+ T cells, as well as a dramatic reduction in the growth of experimental tumors in comparison to wild-type controls. A2a adenosine receptor signaling has also been implicated in adenosine-mediated inhibition of cytokine production and cytotoxic activity by activated natural killer (NK) cells, although the process of NK cell granule exocytosis is apparently suppressed via a distinct and as yet uncharacterized adenosine receptor. In this report, we review the evidence that adenosine is a potent inhibitor of cellular immune responses and may therefore be a major barrier to the successful immunotherapy of human carcinomas. The signaling pathways through which adenosine exerts its inhibitory effects on cell-mediated immune responses are also discussed. The accumulated evidence suggests that the effectiveness of immune-based therapies for solid tumors may be enhanced by selective antagonism of the adenosine receptor subtypes through which adenosine inhibits the anti-tumor activity of T lymphocytes and NK cells.


Assuntos
Adenosina/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Evasão Tumoral/efeitos dos fármacos , Adenosina/metabolismo , Adenosina/fisiologia , Adenosina Desaminase/metabolismo , Adenosina Desaminase/fisiologia , Animais , Humanos , Imunoterapia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Modelos Biológicos , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/terapia , Receptores Purinérgicos P1/metabolismo , Receptores Purinérgicos P1/fisiologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
12.
Oncol Rep ; 19(6): 1511-6, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18497958

RESUMO

Pharmacological inhibitors of the human ether-a-go-go (hEAG) potassium channel, astemizole and imipramine, have been used to demonstrate that hEAG plays a role in cancer cell proliferation. Astemizole and imipramine are, however, relatively non-specific ion channel blockers, as astemizole can also block the related potassium channel, human ether-a-go-go-related (hERG). Therefore, we aimed to determine the molecular target of astemizole, in the human mammary carcinoma cell line MCF-7. We initially confirmed the expression of KCNH1 and KCNH2 mRNA and hEAG and hERG channel protein in MCF-7 cells. Using a [3H]-thymidine incorporation assay we determined that astemizole inhibited MCF-7 cell proliferation, whereas the hERG-specific channel blocker E-4031 had no effect. We then determined that E-4031 inhibited the regulatory volume decrease (RVD) observed in these cells following exposure to hypotonic solutions, confirming that functional hERG channels are present and may be important for cell volume regulation in MCF-7 cells. Our results suggest, for the first time, that hERG is involved in cell volume regulation. In addition, the function of hEAG and hERG in MCF-7 cell proliferation can be separated pharmacologically by utilizing the channel inhibitors astemizole and E-4031. The hEAG channel function in MCF-7 cells appears to be involved in the regulation of cell proliferation, whereas hERG is involved in cell volume regulation.


Assuntos
Neoplasias da Mama/metabolismo , Canais de Potássio Éter-A-Go-Go/fisiologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Antialérgicos/farmacologia , Antiarrítmicos/farmacologia , Astemizol/farmacologia , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Humanos , Imipramina/farmacologia , Síndrome do QT Longo , Piperidinas/farmacologia , Piridinas/farmacologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
13.
Mol Carcinog ; 46(12): 971-80, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17477358

RESUMO

Inhibition of cyclooxygenase (COX)-2 is reported to suppress growth and induce apoptosis in human esophageal adenocarcinoma (EADC) cells, although the precise biologic mechanism is unclear. In this study we tested the hypothesis that the antitumor activity of COX-2 inhibitors may involve modulation of basic fibroblast growth factor (FGF-2), which is overexpressed in EADC. We evaluated the effects of NS-398, a selective COX-2 inhibitor, on FGF-2 expression and proliferation of EADC cell lines that express COX-2 and those that do not. We also correlated COX-2 and FGF-2 expression with clinico-pathologic findings and outcome in a well-characterized series of surgically resected EADC tissues. Seg-1 cells robustly expressed COX-2 and FGF-2, whereas Bic-1 cells expressed neither transcript. FGF-2 was reduced to undetectable levels in Seg-1 cells following NS-398 treatment, but increased within 4 h of drug removal. NS-398 significantly inhibited the growth of Seg-1 cells, and this effect was ameliorated by addition of exogenous FGF-2. In contrast, NS-398 had no effect on Bic-1 cell proliferation and FGF-2 alone had no effect on proliferation of either cell line. NS-398, or a neutralizing anti-FGF-2 antibody, induced apoptosis in Seg-1 cells, and these effects were inhibited by addition of exogenous FGF-2. COX-2 protein was strongly expressed in 46% (10/22) of EADCs, and was associated with a trend towards reduced disease-free survival. These findings indicate that the antitumor effects of COX-2 inhibition in EADC cells may be mediated via suppression of FGF-2, and that COX-2 may be a clinically relevant molecular marker in the management of human EADC.


Assuntos
Adenocarcinoma/enzimologia , Ciclo-Oxigenase 2/química , Inibidores de Ciclo-Oxigenase/farmacologia , Neoplasias Esofágicas/enzimologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Nitrobenzenos/farmacologia , Sulfonamidas/farmacologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Idoso , Apoptose , Western Blotting , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ciclo-Oxigenase 2/biossíntese , Ciclo-Oxigenase 2/genética , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/patologia , Feminino , Fator 2 de Crescimento de Fibroblastos/genética , Imunofluorescência , Humanos , Metástase Linfática/patologia , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas/efeitos dos fármacos
14.
Int J Oncol ; 30(5): 1215-22, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17390024

RESUMO

Peroxisome proliferator activated receptor (PPAR) gamma is a nuclear receptor involved primarily in lipid and glucose metabolism. PPARgamma is also expressed in several cancer types, and has been suggested to play a role in tumor progression. PPARgamma agonists have been shown to reduce the growth of colorectal carcinoma cells in culture and in xenograft models. Furthermore, the PPARgamma agonist thiazolidinedione has been shown to reduce metastasis in a murine model of rectal cancer. Since the chemokine receptor CXCR4 has emerged as an important player in tumorigenesis, particularly in the process of metastasis, we sought to determine if PPARgamma agonists might act in part by reducing CXCR4 expression. We found that rosiglitazone, a thiazolidinedione PPARgamma agonist used primarily in the treatment of type 2 diabetes, significantly reduced cell-surface expression of CXCR4 protein on HT-29 human colorectal carcinoma cells. This effect occurred at concentrations as low as 1 nM, and was first evident after 8 h of drug exposure. CXCR4 mRNA was also down-regulated after treatment with rosiglitazone, indicating that the effect occurs at the level of transcription. Four other thiazolidinedione compounds (ciglitazone, pioglitazone, troglitazone, and MCC555) also significantly reduced CXCR4 expression. To confirm the involvement of PPARgamma in thiazolidinedione-induced CXCR4 down-regulation, we used PPARgamma antagonists GW9662 and T0070907, both of which completely blocked the effect of rosiglitazone on CXCR4 expression. Furthermore, HT-29 cells in which PPARgamma expression was reduced using shRNA were less responsive to rosiglitazone. In conclusion, we have shown that thiazolidinedione compounds reduce CXCR4 mRNA and cell-surface protein expression in a PPARgamma-dependent manner.


Assuntos
Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica , PPAR gama/metabolismo , Receptores CXCR4/genética , Tiazolidinedionas/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Relação Dose-Resposta a Droga , Regulação para Baixo , Humanos , Pioglitazona , Rosiglitazona
15.
Oncogene ; 24(54): 8012-24, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16158060

RESUMO

TGF-beta/Smads regulate a wide variety of biological responses through transcriptional regulation of target genes. ELF, a beta-spectrin, plays a key role in the transmission of TGF-beta-mediated transcriptional response through Smads. ELF was originally identified as a key protein involved in endodermal stem/progenitor cells committed to foregut lineage. Also, as a major dynamic adaptor and scaffolding protein, ELF is important for the generation of functionally distinct membranes, protein sorting and the development of polarized differentiated epithelial cells. Disruption of elf results in the loss of Smad3/Smad4 activation and, therefore, a disruption of the TGF-beta pathway. These observations led us to pursue the function of ELF in gastrointestinal (GI) epithelial cell-cell adhesion and tumor suppression. Here, we show a significant loss of ELF and reduced Smad4 expression in human gastric cancer tissue samples. Also, of the six human gastric cancer cell lines examined, three show deficient ELF expression. Furthermore, we demonstrate the rescue of E-cadherin-dependent homophilic cell-cell adhesion by ectopic expression of full-length elf. Our results suggest that ELF has an essential role in tumor suppression in GI cancers.


Assuntos
Efrina-A2/metabolismo , Neoplasias Gastrointestinais/metabolismo , Neoplasias Gastrointestinais/patologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Anticorpos Monoclonais/metabolismo , Western Blotting , Caderinas/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Neoplasias Gastrointestinais/genética , Humanos , Imuno-Histoquímica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Microscopia Confocal , Testes de Precipitina , Proteína Smad4/metabolismo
16.
Biochem Pharmacol ; 66(9): 1737-47, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14563484

RESUMO

Adenosine is a purine nucleoside which is present at micromolar concentrations in the extracellular fluid of solid cancers as a result of tissue hypoxia. Adenosine acts to promote tumor survival by inhibiting the cell-mediated anti-tumor immune response. However, its role in modulating proliferation of the tumor cell population is unclear. Differing results have been obtained using adenosine analogues or by interfering with adenosine metabolism. We examined the effect of adenosine itself on DNA synthesis and cell growth in six different human and mouse colorectal carcinoma cell lines, from different sites and at different stages of differentiation. Adenosine given as a single dose consistently stimulated DNA synthesis and cell proliferation in all cell lines tested, with an EC(50) of 3.8-30 microM and a maximum stimulation being reached at 10-100 microM. AMP and ATP also stimulated cell proliferation at similar doses. The stimulation by adenosine varied depending upon the culture cell density, with the greatest mitogenic effect at subconfluent densities. Adenosine was metabolized by cellular adenosine deaminase and adenosine kinase. The half-life (t(1/2)) for the decline in adenosine concentration in the medium following a single addition was between 40 min and 3 hr depending on the cell line and culture conditions. The rate of production of endogenous adenosine was low under normoxic culture conditions. Continuous dosing of cultures with adenosine to provide a steady-state concentration showed that proliferation could be stimulated by low micromolar concentrations of adenosine. We conclude that adenosine is stimulatory to the growth of human colorectal carcinoma cells at concentrations present within the tumor extracellular environment.


Assuntos
Adenosina/farmacologia , Adenosina/metabolismo , Adenosina Desaminase/metabolismo , Monofosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Contagem de Células , Divisão Celular/efeitos dos fármacos , Neoplasias Colorretais/patologia , Meios de Cultura/química , DNA/biossíntese , DNA/efeitos dos fármacos , Humanos , Células Tumorais Cultivadas
17.
Anticancer Res ; 22(2A): 673-6, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12014636

RESUMO

BACKGROUND: Although chemotherapeutic agents are known to sensitize some tumour types to killing through cell surface death receptors for Fas ligand and tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), chemosensitization of breast carcinoma cells has not yet been explored. MATERIALS AND METHODS: Mitochondrial thiazole tetrazolium assays were used to measure changes in MCF-7 and T-47D breast carcinoma cell viability. Semiquantitative RT-PCR was used to determine Fas and TRAIL receptor mRNA expression. RESULTS: Treatment with suboptimal concentrations of etoposide or doxorubicin rendered T-47D cells sensitive to anti-Fas antibody or TRAIL, consistent with Fas and TRAIL-R1 mRNA expression by T-47D cells following drug treatment. In contrast, neither drug sensitized MCF-7 cells to TRAIL- or anti-Fas antibody, most likely due to diminished Fas and TRAIL-R1 expression following drug treatment. CONCLUSION: These findings suggest that some breast carcinomas may respond favorably to a combination of chemotherapy and immunotherapy.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Apoptose/fisiologia , Neoplasias da Mama/tratamento farmacológico , Glicoproteínas de Membrana/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Receptor fas/fisiologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Neoplasias da Mama/patologia , Doxorrubicina/farmacologia , Sinergismo Farmacológico , Etoposídeo/farmacologia , Humanos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas Recombinantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas , Receptor fas/biossíntese , Receptor fas/genética , Receptor fas/imunologia
18.
In Vitro Cell Dev Biol Anim ; 39(8-9): 340-2, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14640787

RESUMO

The cytotoxic activity of short-chain (C(2)) ceramide was evaluated in human intestinal carcinoma cells grown as multicellular tumor spheroids versus the same cells cultured as monolayers under closely comparable conditions. A decrease in cell number was seen in monolayer cultures of HT-29, Caco-2, and HRT-18 cells, with an EC(50) (concentration for half-maximal toxicity) of between 13 and 23 microM. However, when the same cells were grown in the multicellular spheroid format, C(2) was markedly less potent in reducing cell number, with an EC(50) of between 44 and 63 microM, representing a 1.9- to 4.9-fold decrease in its potency. The chemotherapeutic agents 5-fluorouracil and cisplatin were equally potent against spheroids and monolayer cultures, indicating that although drug access is a problem in conventionally grown tumor spheroids it is not a problem for spheroids grown under the conditions used in this study. Our results suggest that although ceramide is capable of inducing cell death in intestinal carcinoma cells grown in spheroid culture, its cellular toxicity is constrained by influences that are independent of drug access and may be the consequence of the altered cellular relationships. Carcinoma cell populations show an intrinsically decreased responsiveness to the effects of ceramide when they are grown in a three-dimensional culture format.


Assuntos
Antineoplásicos/toxicidade , Carcinoma/metabolismo , Técnicas de Cultura de Células/métodos , Ceramidas/toxicidade , Neoplasias Intestinais/metabolismo , Esferoides Celulares/metabolismo , Células Tumorais Cultivadas/metabolismo , Antineoplásicos/uso terapêutico , Carcinoma/tratamento farmacológico , Carcinoma/patologia , Ceramidas/química , Ceramidas/uso terapêutico , Cisplatino/uso terapêutico , Cisplatino/toxicidade , Fluoruracila/uso terapêutico , Fluoruracila/toxicidade , Humanos , Neoplasias Intestinais/tratamento farmacológico , Neoplasias Intestinais/patologia , Esferoides Celulares/citologia , Esferoides Celulares/efeitos dos fármacos , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/efeitos dos fármacos
19.
Mol Nutr Food Res ; 57(1): 126-44, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23197449

RESUMO

Apigenin (4',5,7-trihydroxyflavone, 5,7-dihydroxy-2-(4-hydroxyphenyl)-4H-1-benzopyran-4-one) is a flavonoid found in many fruits, vegetables, and herbs, the most abundant sources being the leafy herb parsley and dried flowers of chamomile. Present in dietary sources as a glycoside, it is cleaved in the gastrointestinal lumen to be absorbed and distributed as apigenin itself. For this reason, the epithelium of the gastrointestinal tract is exposed to higher concentrations of apigenin than tissues at other locations. This would also be true for epithelial cancers of the gastrointestinal tract. We consider the evidence for actions of apigenin that might hinder the ability of gastrointestinal cancers to progress and spread. Apigenin has been shown to inhibit cell growth, sensitize cancer cells to elimination by apoptosis, and hinder the development of blood vessels to serve the growing tumor. It also has actions that alter the relationship of the cancer cells with their microenvironment. Apigenin is able to reduce cancer cell glucose uptake, inhibit remodeling of the extracellular matrix, inhibit cell adhesion molecules that participate in cancer progression, and oppose chemokine signaling pathways that direct the course of metastasis into other locations. As such, apigenin may provide some additional benefit beyond existing drugs in slowing the emergence of metastatic disease.


Assuntos
Apigenina/farmacologia , Neoplasias Gastrointestinais/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Frutas/química , Humanos , Extratos Vegetais/farmacologia , Plantas Medicinais/química , Verduras/química
20.
Clin Exp Metastasis ; 28(4): 337-49, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21298326

RESUMO

There is accumulating evidence that secondary plant metabolites such as flavonoids may have anti-cancer properties, and yet the molecular pathways that lead to alterations in cancer cell behaviour remain unclear. We investigated the possible actions of apigenin, a flavone present in leafy vegetables like parsley, on the levels of CD26 in carcinoma cells. CD26 is a multifunctional cell-surface protein that through its associated dipeptidyl peptidase (DPPIV) and ecto-adenosine deaminase (eADA) enzyme activities is able to suppress pathways involved in tumour metastasis. CD26 is down-regulated in various cancers including colorectal carcinoma. Apigenin substantially up-regulated cell-surface CD26 on HT-29 and HRT-18 human colorectal cancer cells. Levels of CD26 protein, along with its associated DPPIV enzyme activity, capacity to bind eADA, and ability to link cells to fibronectin, were increased with a maximum after 24-48 h. Elevation of CD26 occurred at concentrations that were at least 10-fold less than those shown to affect cell growth, and 100-fold below those that could affect cell viability. Furthermore, the CD26 effect was enhanced when apigenin was paired with chemotherapeutic agents utilized in the treatment of advanced colorectal cancer including irinotecan, 5-fluorouracil and oxaliplatin. For irinotecan, apigenin caused a 4-fold increase in the potency of the drug. These results demonstrate that apigenin can increase the cellular levels of CD26 and its multiple functions, and may oppose the predicted effect of decreased DPPIV and eADA activities on carcinoma cells, which is to facilitate tumour growth and metastasis.


Assuntos
Apigenina/farmacologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Dipeptidil Peptidase 4/metabolismo , Metástase Neoplásica/tratamento farmacológico , Adenosina Desaminase/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA