Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Pharmacokinet Pharmacodyn ; 36(6): 523-39, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19904585

RESUMO

This report generates efficient experimental designs (dose, sampling times) for parameter estimation for four basic physiologic indirect pharmacodynamic response (IDR) models. The principles underlying IDR models and their response patterns have been well described. Each IDR model explicitly contains four parameters, k (in) (production), k (out) (loss), I (max)/S (max) (capacity) and IC (50)/SC (50) (sensitivity). The pharmacokinetics of an IV dose of drug described by a monoexponential function of time with two parameters, V and k (el), is assumed. The random errors in the response variable are assumed to be additive, independent, and normal with zero mean and variance proportional to some power of the mean response. Optimal design theory was used extensively to assess the role of both dose and sampling times. Our designs were generated in Mathematica (ADAPT 5 typically produces identical results). G-optimality was used to verify that the generated designs were indeed D-optimal. Such designs are efficient and robust when good prior knowledge of the estimated parameters is available. The efficiency of unconstrained D-optimal designs (4 dose, sampling time pairs) does not improve much when the drug doses are allowed to differ, compared with constrained single dose designs (4 sampling times) with one maximal feasible dose. Also, explored were efficiencies of alternative study designs and results from parameter misspecification. This analysis substantiates the importance of larger doses yielding greater certainty in parameter estimation in pharmacodynamics.


Assuntos
Modelos Biológicos , Farmacocinética , Farmacologia , Animais , Simulação por Computador , Relação Dose-Resposta a Droga , Cálculos da Dosagem de Medicamento , Humanos , Software
2.
Cancer Genomics Proteomics ; 5(1): 43-53, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18359979

RESUMO

BACKGROUND: Time-course and concentration-effect experiments with multiple time-points and drug concentrations provide far more valuable information than experiments with just two design-points (treated vs. control), as commonly performed in most microarray studies. Analysis of the data from such complex experiments, however, remains a challenge. MATERIALS AND METHODS: Here we present a semi-automated method for fitting time profiles and concentration-effect patterns, simultaneously, to gene expression data. The submodels for time-course included exponential increase and decrease models with parameters, such as initial expression level, maximum effect, and rate-constant (or half-time). The submodel for concentration-effect was a 4-parameter Hill model. RESULTS: The method was applied to an Affymetrix HG-U95Av2 dataset consisting of 51 arrays. The specific study focused on the effects of two platinum drugs, cisplatin and oxaliplatin, on A2780 human ovarian carcinoma cells. Replicates were available at most time points and concentrations. Eighteen genes were selected, and after selection, time-course and concentration-effect were modeled simultaneously. CONCLUSION: Comparisons of model parameters helped to distinguish genes with different expression patterns between the two drug treatments. This overall paradigm can help in understanding the molecular mechanisms of the agents, and the timing of their actions.


Assuntos
Antineoplásicos/farmacologia , Perfilação da Expressão Gênica , Expressão Gênica/efeitos dos fármacos , Genes Neoplásicos , Análise de Sequência com Séries de Oligonucleotídeos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Relação Dose-Resposta a Droga , Humanos , Cinética , Modelos Estatísticos , Compostos Organoplatínicos/farmacologia , Oxaliplatina , Análise de Regressão
3.
Cancer Chemother Pharmacol ; 59(6): 711-23, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17021820

RESUMO

PURPOSE: As a follow-up to our previous findings that platinum drugs induce a key enzyme in polyamine catabolism, gene expression profiling and mathematical modeling were used to define the effects of cisplatin and oxaliplatin on the expression of polyamine metabolic pathway genes in A2780 human ovarian carcinoma cells. METHODS: Time-course and concentration-effect experiments were each carried out with cisplatin or oxaliplatin in two separate experiments and cells subjected to gene expression profiling using Affymetrix array technology. Time-course data were modeled using exponential increase and decrease models. Concentration-effect data were modeled using a four parameter Hill model. RESULTS: Gene expression profiling of human ovarian carcinoma A2780 cells after exposure to either cisplatin or oxaliplatin indicates that the expression of several genes involved in polyamine pathway is affected by the platinum drugs. Mathematical/Statistical modeling of the data from time-course and concentration-effect experiments of gene expression from nine polyamine pathway genes represented on the HGU95Av2 chip, indicates that three biosynthetic pathway genes (SAMDC, ODC1 and SRM) are down-regulated and one catabolic pathway gene (SSAT) is up-regulated. Expression changes were similar for different probesets for a given gene on the array. Studies on the induction of SSAT by platinum drugs suggested by the Affymetrix data have been previously validated from this laboratory (Hector et al. in Mol Cancer Ther 3:813-822, 2004). Here, the effects of oxaliplatin exposure on SAMDC and ODC observed by Affymetix are validated with real time QRT-PCR. CONCLUSION: The data indicate a concerted effect of platinum drugs on the polyamine metabolic pathway with down-regulation in the expression of several enzyme genes involved in biosynthesis and many-fold up-regulation in expression of SSAT, an acetylating enzyme gene that is critically involved in polyamine catabolism and export.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Perfilação da Expressão Gênica , Compostos Organoplatínicos/farmacologia , Neoplasias Ovarianas/genética , Aciltransferases/genética , Relação Dose-Resposta a Droga , Feminino , Seguimentos , Humanos , Metabolismo , Modelos Teóricos , Neoplasias Ovarianas/tratamento farmacológico , Oxaliplatina , Poliaminas/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas
4.
Cancer Chemother Pharmacol ; 57(1): 40-5, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16001178

RESUMO

PURPOSE: To measure skin photosensitivity in cancer patients infused with the new second-generation photodynamic sensitizer Photochlor (2-[1-hexyloxyethyl]-2-devinyl pyropheophorbide-a). A major disadvantage of using the clinically approved photosensitizer Photofrin is potentially prolonged and sometimes severe cutaneous phototoxicity. PATIENTS AND METHODS: Forty-eight patients enrolled in Phases 1 and 2 clinical trials underwent two or more exposures to four graded doses (44.4, 66.6, 88.8 or 133.2 J/cm2) of artificial solar-spectrum light (SSL) before and after administration of Photochlor at a dose of 2.5, 3, 4, 5 or 6 mg/m2 . RESULTS: The most severe skin response, experienced by only six of the subjects, was limited to erythema without edema and could only be elicited by exposure to the highest light dose. Conversely, eight subjects had no discernible reaction to SSL at any light dose. For nearly all the patients, the peak skin response was obtained when the interval between sensitizer injection and exposure to SSL was 1 day and, generally, their sensitivity to SSL decreased with increasing sensitizer-light interval. For example, a 2-day sensitizer-SSL interval resulted in less severe reactions than those obtained with the 1-day interval in 79% of the subjects, while 90% of the subjects exposed to SSL 3 days after Photochlor infusion had responses that were less severe than those obtained with either the 1- or 2-day sensitizer-SSL interval. CONCLUSIONS: Photochlor, at clinically effective antitumor doses, causes only mild skin photosensitivity that declines rapidly over a few days.


Assuntos
Éter de Diematoporfirina/efeitos adversos , Neoplasias/tratamento farmacológico , Fotoquimioterapia , Transtornos de Fotossensibilidade/induzido quimicamente , Fármacos Fotossensibilizantes/efeitos adversos , Éter de Diematoporfirina/uso terapêutico , Feminino , Humanos , Masculino , Fármacos Fotossensibilizantes/uso terapêutico , Pele/efeitos dos fármacos , Pele/efeitos da radiação , Raios Ultravioleta
5.
Cancer Chemother Pharmacol ; 57(1): 73-83, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16010591

RESUMO

PURPOSE: Based on reported synergy of the topoisomerase-I (topo-I) inhibitor irinotecan with antimetabolites, irinotecan and cytarabine (Ara-C) were administered sequentially to patients with acute myeloid leukemia (AML) refractory to or relapsed following high-dose Ara-C and anthracycline therapy. Pharmacokinetic and pharmacodynamic studies were performed with the first irinotecan dose. EXPERIMENTAL DESIGN: In vitro synergy of irinotecan followed by Ara-C was confirmed in a human AML cell line as a basis for the clinical trial. Irinotecan was administered daily for 5 days, with Ara-C 1 g/m2 12 h after each irinotecan dose. Irinotecan was initiated at 5 mg/m2, and the dose was escalated by 5 mg/m2 increments in cohorts of three patients and in individual patients. Pre-treatment samples were studied for topo-I activity and serial samples after the first irinotecan dose were analyzed for pharmacokinetics and for pharmacodynamic effects, including DNA damage and DNA synthesis rate. RESULTS: The irinotecan dose reached 15 mg/m2 in three-patient cohorts without reaching the maximum tolerated dose, and reached 30 mg/m2 in individual patients. The AUC and Cmax of both irinotecan and its active metabolite SN38 increased linearly in proportion to dose, and the mean half-lives of irinotecan conversion to SN38 and SN38 elimination were 6.2 h (CV 171%) and 7.2 h (CV 48%). Irinotecan rapidly induced DNA damage, and DNA synthesis inhibition varied among patients and treatment cycles. All courses resulted in rapid cytoreduction, and two patients achieved complete remission. Topo-I activity did not predict response. CONCLUSION: Irinotecan can be safely administered with Ara-C. This combination is active in refractory AML and warrants further study.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia Mieloide/tratamento farmacológico , Doença Aguda , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Camptotecina/administração & dosagem , Camptotecina/efeitos adversos , Camptotecina/análogos & derivados , Camptotecina/farmacocinética , Camptotecina/uso terapêutico , Citarabina/administração & dosagem , Citarabina/efeitos adversos , Citarabina/farmacocinética , Citarabina/uso terapêutico , Dano ao DNA , DNA Topoisomerases Tipo I/metabolismo , DNA de Neoplasias/biossíntese , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Células HL-60 , Humanos , Irinotecano , Leucemia Mieloide/patologia , Masculino , Pessoa de Meia-Idade , Recidiva
6.
Clin Cancer Res ; 11(6): 2320-6, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15788683

RESUMO

PURPOSE: Overexpression of the multidrug resistance proteins P-glycoprotein (Pgp), multidrug resistance protein (MRP-1), breast cancer resistance protein (BCRP), and lung resistance protein (LRP) is associated with treatment failure in acute myeloid leukemia (AML) and other malignancies. The Pgp modulator cyclosporin A has shown clinical efficacy in AML, whereas its analogue PSC-833 has not. Cyclosporin A is known to also modulate MRP-1, and we hypothesized that broad-spectrum multidrug resistance modulation might contribute to its clinical efficacy. EXPERIMENTAL DESIGN: We studied the effects of cyclosporin A and PSC-833 on in vitro drug retention and cytotoxicity in resistant cell lines overexpressing Pgp, MRP-1, and BCRP and on nuclear-cytoplasmic drug distribution and cytotoxicity in cells overexpressing LRP. Cellular drug content was assessed by flow cytometry and nuclear-cytoplasmic drug distribution by confocal microscopy. RESULTS: Cyclosporin A enhanced retention of the substrate drug mitoxantrone in cells overexpressing Pgp (HL60/VCR), MRP-1 (HL60/ADR), and BCRP (8226/MR20, HEK-293 482R) and increased cytotoxicity 6-, 4-, 4-, and 3-fold, respectively. Moreover, cyclosporin A enhanced nuclear distribution of doxorubicin in 8226/MR20 cells, which also express LRP, and increased doxorubicin cytotoxicity 12-fold without an effect on cellular doxorubicin content, consistent with expression of wild-type BCRP, which does not efflux doxorubicin. Cyclosporin A also enhanced nuclear doxorubicin distribution in a second cell line with LRP overexpression, HT1080/DR4. PSC-833 enhanced mitoxantrone retention and cytotoxicity in cells overexpressing Pgp, but had no effect in cells overexpressing MRP-1, BCRP, or LRP. CONCLUSIONS: Cyclosporin A modulates Pgp, MRP-1, BCRP, and LRP, and this broad-spectrum activity may contribute to its clinical efficacy.


Assuntos
Apoptose/efeitos dos fármacos , Ciclosporina/farmacologia , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Antibióticos Antineoplásicos/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Núcleo Celular/metabolismo , Células Cultivadas , Ciclosporina/farmacocinética , Citoplasma/metabolismo , Doxorrubicina/metabolismo , Células HL-60/efeitos dos fármacos , Células HL-60/metabolismo , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Mitoxantrona/metabolismo , Proteínas de Neoplasias/metabolismo , Partículas de Ribonucleoproteínas em Forma de Abóbada/metabolismo
7.
Reg Anesth Pain Med ; 31(1): 14-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16418019

RESUMO

BACKGROUND AND OBJECTIVES: The adrenergic drug ephedrine inhibits Na(+) current in cultured cells expressing Na(+) channels and provides dose-dependent reversible rat sciatic nerve blockade. The dosage required for peripheral nerve blockade in humans would cause unacceptable cardiovascular side effects. We therefore hypothesized that either lidocaine or bupivacaine would show synergy with ephedrine in a rat sciatic nerve block model, allowing a dose reduction and limiting side effects while improving efficacy. METHODS: Sciatic nerves of anesthetized rats were exposed by lateral incision of the thighs; 0.2 mL of different concentrations of the following drugs was injected sub-fascially: bupivacaine, lidocaine, and ephedrine alone and bupivacaine or lidocaine combined with ephedrine (n=8 per group). After animals recovered from anesthesia, a blinded investigator evaluated motor blockade (push against a balance) and sensory/nociceptive blockade (to pinch of the fifth toe) at predefined time intervals. RESULTS: Ephedrine combined with bupivacaine interacted additively for both motor and sensory blockade, and ephedrine combined with lidocaine interacted antagonistically (sub-additive) for sensory blockade and additively for motor blockade. CONCLUSIONS: A combination of ephedrine with either lidocaine or bupivacaine is unlikely to allow a significant dose reduction because of a lack of synergy. Furthermore, the cardiovascular side effects will limit the maximal tolerable dosage of ephedrine.


Assuntos
Adrenérgicos/administração & dosagem , Anestésicos Locais/administração & dosagem , Bupivacaína/administração & dosagem , Efedrina/administração & dosagem , Lidocaína/administração & dosagem , Bloqueio Nervoso , Nervo Isquiático/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Potencial Evocado Motor/efeitos dos fármacos , Potenciais Somatossensoriais Evocados/efeitos dos fármacos , Masculino , Modelos Animais , Medição da Dor , Ratos , Ratos Sprague-Dawley
8.
Cancer Res ; 63(23): 8126-31, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14678965

RESUMO

Using the photosensitizer 2-[1-hexyloxyethyl]-2-devinyl pyropheophorbide-a, we have determined that photodynamic therapy (PDT) can be used to facilitate the delivery of macromolecular agents. PDT regimens that use low fluences and fluence rates were the most successful. This effect was demonstrated for fluorescent microspheres with diameters ranging from 0.1 to 2 microm. Such treatment given immediately before administration of Doxil, a liposomally encapsulated formulation of doxorubicin with an average diameter of 0.1 microm, significantly enhanced its accumulation in transplanted murine Colo 26 tumors. The combination of PDT and Doxil led to a highly significant potentiation in tumor control without concomitant enhancement of systemic or local toxicity. Interestingly, concentration-effect modeling suggested that the enhanced cure rate was greater than what was predicted based on the increase in intratumor Doxil concentration. In summary, we have developed a novel PDT treatment that enhances the delivery and efficacy of macromolecule-based cancer therapies such as Doxil.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Clorofila/análogos & derivados , Clorofila/farmacologia , Doxorrubicina/administração & dosagem , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Animais , Antibióticos Antineoplásicos/farmacocinética , Permeabilidade Capilar/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Terapia Combinada , Doxorrubicina/farmacocinética , Sistemas de Liberação de Medicamentos , Sinergismo Farmacológico , Feminino , Camundongos , Camundongos Endogâmicos BALB C
9.
Cancer Res ; 63(22): 7584-90, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14633671

RESUMO

DMXAA (5,6-dimethylxanthenone-4-acetic acid) is an antivascular agent that exerts its antitumor effect at least partly through the induction of tumor necrosis factor (TNF)-alpha. Photodynamic therapy (PDT), the activation of a photoreactive drug in tumor tissue with visible light, is used clinically to control solid malignancies. PDT has been shown previously to be potentiated, in mice, by the i.p. administration of recombinant human TNF-alpha. Here, we investigated the activity of DMXAA as a modifier of Photofrin-based PDT of implanted murine RIF-1 tumors. The DMXAA dose (20 mg.kg(-1)) used throughout this study had little effect on tumor growth. The combination of DMXAA and PDT led to a reduction in tumor volume and significant delays in regrowth, giving a PDT-dose modification factor of 2.81. This enhancement was found to be strongly schedule dependent. The most pronounced responses were achieved when DMXAA was administered 1-3 h before the local illumination of the tumors; less activity was observed at other intervals within +/-24 h of PDT-light delivery. Using a 2-h DMXAA-light interval, histological examination showed significantly reduced blood vessel counts (CD31 immunostaining) and marked necrosis (H&E) in the tumors given combination therapy compared with the tumors given either agent alone. Conversely, peritumoral tissue was still intact 24 h after the combined therapy. DMXAA did not augment the damage to normal mouse feet after low-dose PDT (1.5 mg.kg(-1) Photofrin); however, there was some enhancement of normal tissue phototoxicity when DMXAA was combined with high-dose PDT. The antitumor effect after DMXAA plus low-dose PDT (1.5 mg.kg(-1) Photofrin) appeared to be dependent on TNF-alpha because neutralizing antibodies to this cytokine reduced the tumor response to control levels. DMXAA by itself induced TNF-alpha in RIF-1 tumors whereas PDT did not. However, the addition of PDT after DMXAA resulted in decreases in TNF-alpha, suggesting that the enhanced antitumor activity of the combination therapy was not attributable simply to an increased induction of the cytokine by PDT over that from DMXAA alone. These observations suggest a promising new combination therapy with considerable therapeutic advantage.


Assuntos
Neoplasias Experimentais/tratamento farmacológico , Fotoquimioterapia/métodos , Fator de Necrose Tumoral alfa/biossíntese , Xantonas/farmacologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Linhagem Celular Tumoral , Éter de Diematoporfirina/farmacologia , Relação Dose-Resposta a Droga , Esquema de Medicação , Sinergismo Farmacológico , Feminino , Fibrossarcoma/irrigação sanguínea , Fibrossarcoma/tratamento farmacológico , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Camundongos , Camundongos Endogâmicos C3H , Necrose , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Fator de Necrose Tumoral alfa/imunologia , Xantonas/antagonistas & inibidores
10.
Cancer Res ; 63(8): 1806-13, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12702566

RESUMO

Photodynamic therapy is an effective and often curative treatment for certain solid tumors. The porphyrin-based photosensitizer Photofrin, the only Food and Drug Administration-approved drug for this therapy, suffers from certain disadvantages: its complex chemical nature; retention by skin (leading to protracted cutaneous photosensitivity); and less than optimal photophysical properties. In this study, we examine the population pharmacokinetics and cutaneous phototoxicity of 2-[1-hexyloxyethyl]-2-devinyl pyropheophorbide-a (HPPH), a chlorin-type photosensitizer with more favorable photophysical properties. HPPH plasma concentration-time data were obtained in 25 patients enrolled in Phase I-II clinical trials for the treatment of partially obstructive esophageal carcinoma, high-grade dysplasia associated with Barrett's esophagus, carcinoma of the lung, or multiple basal cell carcinomas. Doses of 3, 4, 5, or 6 mg/m(2) were administered as 1-h i.v. infusions. The pharmacokinetic data for each patient were fitted with a standard two-compartment (biexponential) model with continuous infusion. The model fitting approach was iteratively reweighted nonlinear regression, with weights equal to the reciprocal of the square of the predicted HPPH plasma concentrations. The complete set of data for all 25 patients was then fitted simultaneously with nonlinear mixed effects modeling. Cutaneous phototoxicity responses were determined, as a function of time after HPPH infusion, following exposure to various doses of light from a solar simulator. The estimates of the population mean (variance) for each parameter were as follows: volume of distribution (V(C)), 2.40 liters/m(2) (0.259); steady-state volume (V(SS)), 9.58 liters/m(2) (11.6); systemic clearance (CL), 0.0296 liter/h/m(2) (0.000094); and distributional clearance (CL(D)), 0.144 liter/h/m(2) (0.00166). These parameters were independent of dose. Clearance increased with age. A relative error model was used for the difference in the raw and fitted data, and the overall coefficient of variation estimate across all of the data was 14.5%. The estimated mean population alpha and beta half-lives (95% confidence interval) were 7.77 h (3.46-17.6 h) and 596 h (120-2951 h), respectively. High-performance liquid chromatography analysis of serum showed no circulating HPPH metabolites, and in vitro incubation of HPPH with human liver microsomal preparations resulted in no metabolite or glucuronic acid-HPPH conjugate production. A minimal skin response to the solar simulator was observed, mostly in patients treated with the highest dose of HPPH, 6 mg/m(2). All of the HPPH pharmacokinetic parameters were consistent with a highly lipophilic agent that is concentrated in plasma and is nearly 100% bound to plasma proteins; this was verified by plasma protein binding studies. Whereas low concentrations of HPPH can be detected in plasma several months after a single infusion, no instances of cutaneous photosensitivity have been noted in these patients. In general, HPPH pharmacokinetic profiles are readily predictable from the global population model. This is the first comprehensive human population pharmacokinetic/pharmacodynamic study of a clinical anticancer photodynamic therapy agent.


Assuntos
Clorofila/análogos & derivados , Clorofila/farmacocinética , Neoplasias/metabolismo , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacocinética , Adulto , Idoso , Idoso de 80 Anos ou mais , Esôfago de Barrett/sangue , Esôfago de Barrett/tratamento farmacológico , Esôfago de Barrett/metabolismo , Proteínas Sanguíneas/metabolismo , Carcinoma Basocelular/sangue , Carcinoma Basocelular/tratamento farmacológico , Carcinoma Basocelular/metabolismo , Clorofila/efeitos adversos , Clorofila/sangue , Clorofila/uso terapêutico , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Neoplasias Esofágicas/sangue , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/metabolismo , Humanos , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Pessoa de Meia-Idade , Neoplasias/sangue , Neoplasias/tratamento farmacológico , Fármacos Fotossensibilizantes/efeitos adversos , Fármacos Fotossensibilizantes/sangue , Fármacos Fotossensibilizantes/uso terapêutico , Pele/efeitos dos fármacos , Neoplasias Cutâneas/sangue , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/metabolismo
11.
Oncol Rep ; 14(4): 925-32, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16142353

RESUMO

The efficacy of platinum drugs in the treatment of cancer is often restricted by the acquisition of tumor cell resistance subsequent to treatment. To better understand mechanisms involved in this phenomenon, a clonal subline (A2780/C10B) isolated from an oxaliplatin-resistant human ovarian carcinoma cell line (A2780/C10) was developed, as reported previously. This cell line is 18-fold resistant to oxaliplatin and shows a 3-fold cross resistance to cisplatin. Here, we report on the gene expression analysis using Affymetrix HG-U95Av2 oligonucleotide arrays of cells in log phase growth from both the parental cell line and drug-resistant variant. Probe level analysis was perfomed using the model based expression index (dChip) and robust multichip average (RMA) methods. Genes that were differentially expressed between the two groups were identified using the significance analysis of microarrays (SAM) method with a minimum false discovery rate <1%. We identified 43 genes that were overexpressed, and 39 underexpressed in the drug-resistant cell line. Collagen VI (COL6A3) was overexpressed 62-fold and the most highly up-regulated gene. This finding is consistent with other published data based on serial analysis of gene expression (SAGE) profiling of cisplatin-resistant and sensitive ovarian carcinoma cells. Among the significant functional groups of overexpressed genes in our study were extracellular matrix genes (9 of 43) and those involved in signal transduction (7 of 43). Extracellular matrix genes included two matrix metalloproteinases (MMP3 and MMP12). Integrin alpha 1 (ITGA1) and WNT5A were also overexpressed. Genes that encode for extracellular matrix proteins were also among those found down-regulated in the resistant cell line. Several genes involved in the regulation of cell cycle and growth were found to be underexpressed, including the suppressor of cytokine signaling 2 (SOCS2), necdin (NDN), and glypicans (GPC3 and GPC4). The mRNA levels of six differentially expressed genes (COL6A3, MMP12, MMP3, WNT5A, NID, and HMGB2) were validated using real-time quantitative RT-PCR. The identification of these genes should aid in a better understanding of the pathways resulting in platinum drug resistance.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Análise de Sequência com Séries de Oligonucleotídeos , Compostos Organoplatínicos/farmacologia , Neoplasias Ovarianas/genética , Linhagem Celular Tumoral , Cisplatino/farmacologia , Colágeno Tipo VI/metabolismo , DNA/química , Reparo do DNA , Matriz Extracelular/metabolismo , Feminino , Proteína HMGB2/biossíntese , Humanos , Integrina alfa1/metabolismo , Metaloproteinase 12 da Matriz , Metaloproteinase 3 da Matriz/biossíntese , Metaloproteinase 3 da Matriz/metabolismo , Glicoproteínas de Membrana/biossíntese , Metaloendopeptidases/biossíntese , Metaloendopeptidases/metabolismo , Sondas de Oligonucleotídeos/química , Oligonucleotídeos/química , Oxaliplatina , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Proteínas Wnt/biossíntese , Proteínas Wnt/metabolismo , Proteína Wnt-5a
12.
Curr Drug Metab ; 4(5): 399-409, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14529372

RESUMO

A flexible approach to response surface modeling for the study of the joint action of three active anticancer agents is used to model a complex pattern of synergism, additivity and antagonism in an in vitro cell growth assay. The method for determining a useful nonlinear response surface model depends upon a series of steps using appropriate scaling of drug concentrations and effects, raw data modeling, and hierarchical parameter modeling. The method is applied to a very large in vitro study of the combined effect of Trimetrexate (TMQ), LY309887 (LY), and Tomudex (TDX) on inhibition of cancer cell growth. The base model employed for modeling dose-response effect is the four parameter Hill equation [1]. In the hierarchical aspect of the final model, the base Hill model is treated as a function of the total amount of the three drug mixture and the Hill parameters, background B, dose for 50% effect D50, and slope m, are understood as functions of the three drug fractions. The parameters are modeled using the canonical mixture polynomials from the mixture experiment methodologies introduced by Scheff [2]. We label the model generated a Nonlinear Mixture Amount model with control observations, or zero amounts, an "NLMAZ" model. This modeling paradigm provides for the first time an effective statistical approach to modeling complex patterns of local synergism, additivity, and antagonism in the same data set, the possibility of including additional experimental components beyond those in the mixture, and the capability of modeling three or more drugs.


Assuntos
Antineoplásicos/farmacologia , Sinergismo Farmacológico , Adenocarcinoma/enzimologia , Algoritmos , Antimetabólitos/química , Antimetabólitos/farmacologia , Divisão Celular , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Hidroximetil e Formil Transferases/antagonistas & inibidores , Neoplasias Intestinais/enzimologia , Modelos Biológicos , Dinâmica não Linear , Fosforribosilglicinamido Formiltransferase , Quinazolinas/química , Quinazolinas/farmacologia , Tetra-Hidrofolatos/química , Tetra-Hidrofolatos/farmacologia , Tiofenos/química , Tiofenos/farmacologia , Trimetrexato/química , Trimetrexato/farmacologia
13.
Front Biosci ; 7: e48-57, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11815284

RESUMO

Use of vaccines to prevent and treat breast and ovarian cancer is a highly attractive approach because of the expected minimal side effects and the potential to predict individuals likely to benefit from vaccination. To fully harness the capacity of the immune system for this purpose, it is necessary to characterize tumor antigens for these cancers so that purified antigens can be tested for their immunogenicity in individual patients and for their suitability as targets of vaccine-induced immunity. Discovery of novel breast and ovarian tumor antigens is also necessary for developing multi-antigen vaccines composed of multiple tumor antigens. Such vaccines are expected to induce diverse immune responses and minimize emergence of antigen-loss variant tumors that are resistant to vaccine-induced immunity. With the exception of melanomas, for most human cancers including breast and ovarian cancers the repertoire of known tumor antigens remains relatively small. In this review we will discuss the importance of characterizing tumor antigens for use in vaccination against cancer and then summarize antigens that have been characterized for human breast and ovarian cancers. We will also emphasize that identification of a novel tumor antigen, while an important first step, needs to be followed by a multi-step process of validation of that antigen. The steps in this validation process are i) to demonstrate that a tumor antigen is over-expressed at a reasonable frequency in primary tumors and in metastases; ii) to demonstrate the immunogenicity of a tumor antigen in an appropriate animal model; iii) to demonstrate its immunogenicity and safety in humans. Additional considerations in this review include: i) discussion of the potential of breast and ovarian tumor antigens as markers for early detection and for monitoring tumor burden in cancer patients; ii) discussion of their potential as prognostic markers of breast and ovarian cancers; and iii) discussion of a unique class of tumor antigens and markers that induce expression of multiple other tumor antigens and markers. Finally, we will discuss the present evidence for potential for autoimmunity that might accompany antitumor vaccination.


Assuntos
Antígenos de Neoplasias/imunologia , Biomarcadores Tumorais/imunologia , Neoplasias da Mama , Neoplasias Ovarianas , Animais , Células Apresentadoras de Antígenos/imunologia , Antígenos de Neoplasias/análise , Antígenos de Neoplasias/metabolismo , Autoimunidade , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/imunologia , Neoplasias da Mama/prevenção & controle , Vacinas Anticâncer , Feminino , Antígenos H-2/genética , Antígeno HLA-A2/genética , Humanos , Camundongos , Metástase Neoplásica , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/prevenção & controle , Prognóstico , Linfócitos T/imunologia
14.
Leuk Res ; 28(5): 449-55, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15068897

RESUMO

Acute myeloid leukemia (AML) with rearrangement of the core-binding factor (CBF) alpha or beta subunit gene has a favorable prognosis, but CD56 expression in CBFalpha-AML is associated with short disease-free survival. A proposed mechanism is overexpression of the multidrug resistance (MDR) protein P-glycoprotein (Pgp). CD56 expression, Pgp expression and function, and expression of the additional MDR proteins multidrug resistance protein-1 (MRP-1), lung resistance protein (LRP) and breast cancer resistance protein (BCRP) were studied in pretreatment blasts from 25 CBF-AML patients. CD56 expression was frequent in CBFalpha but rare in CBFbeta, and Pgp expression and function were frequent in both subtypes. CD56 expression did not correlate with Pgp expression or function, nor with expression of the other MDR proteins. Treatment failure associated with CD56 expression in CBFalpha-AML is not likely attributable to Pgp.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/análise , Antígeno CD56/análise , Proteínas de Ligação a DNA/genética , Rearranjo Gênico , Leucemia Mieloide Aguda/tratamento farmacológico , Fatores de Transcrição/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/fisiologia , Adulto , Subunidades alfa de Fatores de Ligação ao Core , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Humanos , Cariotipagem , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo
15.
Int J Oncol ; 23(2): 401-9, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12851689

RESUMO

In order to examine the intracellular locus of the folic acid (PteGlu)-enhanced synergies of trimetrexate (TMQ) plus the thymidylate synthase (TS) inhibitor, raltitrexed (RTX), and TMQ plus the glycinamide ribonucleotide formyltransferase (GARFT) inhibitor, AG2034, comprehensive protection studies with thymidine (dThd) and hypoxanthine (HX) were conducted in a 96-well plate cell growth inhibition (sulforhodamine B) assay. Current modeling techniques were extended to characterize these protection patterns involving multiple-agent interaction. Wild-type human ileocecal HCT-8 cells and DW2, a subline deficient in folylpoly-gamma-glutamate synthetase (FPGS) were individually treated for 96 h with TMQ, AG2034 and a 1:1 mixture of TMQ:AG2034 or with TMQ, RTX, and a 1:1 mixture of TMQ:RTX in the presence of PteGlu (2.3 or 40 micro M) and the protection agents (10 micro M dThd and/or 100 micro M HX). Drug treatments were randomly assigned to wells. Both isobols and 3-dimensional concentration-effect surfaces were used to assess the nature and the intensity of drug interactions. The structural Hill model was fitted to data with weighted non-linear regression for most cases. A so-called 'double Hill' model was sometimes more appropriate when a plateau in the middle of the concentration-effect curve was found. In HCT-8 and DW2 cells at 2.3 and 40 micro M PteGlu, inhibition of DHFR by TMQ induced antithymidylate and antipurine effects; AG2034 and RTX selectively inhibited de novo purine or thymidine synthesis, respectively. dThd protection increased the PteGlu-enhancement of the TMQ + AG2034 synergy, whereas HX protection increased the PteGlu-enhancement of the TMQ + RTX synergy. The PteGlu-enhanced synergies of TMQ + AG2034 and TMQ + RTX occur primarily through inhibition of purine synthesis and inhibition of thymidylate synthesis, respectively. These results further substantiate the hypothesis that the nonpolyglutamylatable DHFR inhibitor, TMQ, acts as a modulator by decreasing the protection by PteGlu of cells against the polyglutamylatable AG2034 and RTX.


Assuntos
Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Ácido Fólico/farmacologia , Hipoxantina/farmacologia , Timidina/farmacologia , Antineoplásicos/farmacologia , Neoplasias do Ceco/tratamento farmacológico , Neoplasias do Ceco/metabolismo , Divisão Celular/efeitos dos fármacos , Resistência a Múltiplos Medicamentos , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Glutamatos/efeitos adversos , Hematínicos/farmacologia , Humanos , Hidroximetil e Formil Transferases/antagonistas & inibidores , Hipoxantina/metabolismo , Neoplasias do Íleo/tratamento farmacológico , Neoplasias do Íleo/metabolismo , Peptídeo Sintases/deficiência , Fosforribosilglicinamido Formiltransferase , Pirimidinas/efeitos adversos , Quinazolinas/farmacologia , Tiofenos/farmacologia , Timidina/metabolismo , Timidilato Sintase/antagonistas & inibidores , Trimetrexato/efeitos adversos , Células Tumorais Cultivadas
16.
Photochem Photobiol ; 76(1): 91-7, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12126312

RESUMO

A study has been carried out to define the importance of the peripheral benzodiazepine receptor (PBR) as a binding site for a series of chlorin-type photosensitizers, pyropheophorbide-a ethers, the subject of a previous quantitative structure-activity relationship study by us. The effects of the PBR ligand PK11195 on the photodynamic activity have been determined in vivo for certain members of this series of alkyl-substituted ethers: two of the most active derivatives (hexyl and heptyl), the least active derivative (dodecyl [C12]) and one of intermediate activity (octyl [C8]). The photodynamic therapy (PDT) effect was inhibited by PK11195 for both of the most active derivatives, but no effect on PDT activity was found for the less active C12 or C8 ethers. The inhibitory effects of PK11195 were predicted by the binding of only the active derivatives to the benzodiazepine site on albumin, ie. human serum albumin (HSA)-Site II. Thus, as with certain other types of photosensitizers, it has been demonstrated with this series of pyropheophorbide ethers that in vitro binding to HSA-Site II is a predictor of both optimal in vivo activity and binding to the PBR in vivo.


Assuntos
Clorofila/análogos & derivados , Fotoquimioterapia , Receptores de GABA-A/metabolismo , Animais , Sítios de Ligação , Clorofila/uso terapêutico , Humanos , Técnicas In Vitro , Isoquinolinas/farmacologia , Camundongos , Camundongos Endogâmicos C3H , Fármacos Fotossensibilizantes/uso terapêutico , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/metabolismo , Albumina Sérica/metabolismo
17.
Oncol Rep ; 11(2): 453-8, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14719083

RESUMO

We previously reported that prostate derived Ets transcription factor (PDEF) is a breast tumor-associated molecule. To obtain further insights into PDEF expression in other human tumor types, a cDNA library database from human adult normal and tumor tissues was compiled and searched for PDEF distribution. The results showed that PDEF is present at relative higher frequencies in the cDNA libraries from brain, breast, lung and ovarian tumors in comparison to those from the corresponding normal tissues. RT/PCR analysis of PDEF expression in ovarian tumors confirmed that PDEF is expressed in 36 out of 51 (71%) ovarian tumors. Further comparison of the distribution of PDEF with other widely recognized cancer-associated molecules showed that PDEF has more restricted distributions than Her-2/neu, Bcl-2, survivin or telomerase in cDNA libraries from normal human tissues and more increased distribution than Her-2/neu, CA-125, Bcl-2, survivin and telomerase in cDNA libraries from brain (except survivin), breast, lung and ovarian tumors. These data together show a better tumor-association for PDEF and suggest that PDEF is a more suitable target for developing specific cancer therapies.


Assuntos
Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , DNA Complementar/genética , Bases de Dados Factuais , Feminino , Biblioteca Gênica , Humanos , Masculino , Neoplasias Ovarianas , Ovário , Próstata , Ligação Proteica , Proteínas Tirosina Quinases/isolamento & purificação , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/isolamento & purificação , Proteínas Proto-Oncogênicas c-ets , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/isolamento & purificação
18.
Am J Clin Oncol ; 26(1): 6-11, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12576916

RESUMO

The records of 23 patients diagnosed and treated at the Massachusetts General Hospital for extranodal non-Hodgkin's lymphoma of the paranasal sinus and nasal cavity were reviewed. The majority of patients were Ann Arbor stage I and approximately evenly divided in T1 or T2 (n = 10) and T3 or T4 (n = 13). Eight patients had nasal-type NK/T cell and 15 patients had diffuse large B-cell lymphoma (DLBCL). The patients with nasal-type NK/T cell lymphoma predominately involved the nasal cavity (5/8), whereas the DLBCL more often had the paranasal sinuses as the primary site (12/15). All patients received radiation as part of their treatment. Only three patients received chemotherapy as part of their initial treatment for three cycles using a cyclophosphamide, doxorubicin, vincristine, and prednisone-based regimen. By coincidence, the estimated overall survival (OS) and disease-free survival rates for both 5 and 10 years were all the same for all analyses. The OS for the entire group at 10 years was 78%. Significant prognostic factors were Ann Arbor stage IEA versus IIEA ( p = 0.0001) and T stage with (T1 or T2) versus (T3 or T4) (p = 0.0243). Combining Ann Arbor stage and T stage created a highly significant prognostic variable (IEA & [T1 or T2], IEA & [T3 or T4], IIEA & [T1 or T2], IIEA & [T3 or T4]) at p = 0.0001, regardless of site or histology. Patients with local-regional disease appear to be well controlled with radiation alone, but distant failure remains a problem. A combined-modality approach with local-regional radiation and systemic chemotherapy is recommended for these patients.


Assuntos
Linfoma não Hodgkin , Cavidade Nasal , Neoplasias Nasais , Neoplasias dos Seios Paranasais , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Terapia Combinada , Feminino , Humanos , Linfoma não Hodgkin/tratamento farmacológico , Linfoma não Hodgkin/patologia , Linfoma não Hodgkin/radioterapia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Nasais/tratamento farmacológico , Neoplasias Nasais/patologia , Neoplasias Nasais/radioterapia , Neoplasias dos Seios Paranasais/tratamento farmacológico , Neoplasias dos Seios Paranasais/patologia , Neoplasias dos Seios Paranasais/radioterapia , Prognóstico , Dosagem Radioterapêutica , Estudos Retrospectivos , Análise de Sobrevida , Resultado do Tratamento
20.
Front Biosci (Schol Ed) ; 2(2): 454-67, 2010 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-20036961

RESUMO

Our group recently developed a response-surface modeling paradigm (White et al: Curr Drug Metab 2, 399-409, 2003) and tested its application to both mixtures of anticancer agents and antifungals. This new model is a Hill-type equation, with the slope and potency parameters being functions of the normalized drug ratios, using polynomial expressions. Response surface methods allow one to model and interpret all of the information present in the full concentration-effect data set, to visualize local regions of synergy, additivity and antagonism, and even to quantify the degree of synergy or antagonism, both globally, and across local regions of the response surface. In the present article, we study the effect of changes in the different parameters of the polynomial expressions for two-drug and three-drug mixtures, on the geometrical shapes of several 2-dimensional representations of the 3-dimensional concentration-effect surface. A secondary goal of this report is to compare the mathematical characteristics of the rival White and (Minto et al: Anesthesiol 92, 1603-1616, 2000) modeling paradigms.


Assuntos
Antifúngicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Combinação de Medicamentos , Sinergismo Farmacológico , Modelos Biológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA