Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Ann Oncol ; 19(11): 1903-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18632723

RESUMO

BACKGROUND: The purpose of this study was to evaluate the potential of noncytotoxic doses of suramin to reverse chemotherapy resistance in advanced chemonaive and chemoresistant non-small-cell lung cancer patients. PATIENTS AND METHODS: Patients received paclitaxel (Taxol) (200 mg/m(2)) and carboplatin (area under the concentration-time curve 6 mg/ml/min) every 3 weeks. The total suramin per cycle dose was calculated using a nomogram derived from the preceding phase I trial to obtain the desirable plasma concentration range of 10-50 microM. RESULTS: Thirty-nine response-assessable chemonaive patients (arm A) received 213 cycles. Thirty-eight cycles were administered to 15 patients with demonstrated resistance to paclitaxel and carboplatin (arm B). The pattern/frequency of toxic effects was similar to those expected for paclitaxel/carboplatin, and pharmacokinetic analyses (199 cycles) showed suramin plasma concentrations maintained between 10 and 50 microM in 94% of cycles. In arm A, response evaluation criteria in solid tumors (RECIST) response rate was 36% (95% confidence interval 22% to 54%; two complete, 12 partial); 15 patients (38%) had disease stabilization for > or =4 months; median progression-free survival (intention to treat) was 6.4 months; median overall survival (OS) 10.4 months and 1-year survival rate 38%. In arm B, no RECIST responses occurred; four patients had disease stabilization for > or =4 months; median OS was 132 days and 1-year survival rate 7%. Plasma basic fibroblast growth factor levels were higher in chemopretreated/refractory patients compared with chemonaive patients (P = 0.05). Sequence analysis of the EGFR tyrosine kinase domain in a long-term disease-free survivor revealed an ATP-binding pocket mutation (T790M). CONCLUSIONS: Noncytotoxic suramin did not increase paclitaxel/carboplatin's toxicity and the suramin dose was predicted from clinical parameters. No clinically significant reversal of primary resistance was documented, but a modulatory effect in chemotherapy-naive patients cannot be excluded. Controlled randomization is planned for further evaluation of this treatment strategy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Carboplatina/administração & dosagem , Carboplatina/efeitos adversos , Carboplatina/farmacocinética , Carcinoma Pulmonar de Células não Pequenas/sangue , Carcinoma Pulmonar de Células não Pequenas/patologia , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Feminino , Fator 1 de Crescimento de Fibroblastos/sangue , Fator 2 de Crescimento de Fibroblastos/sangue , Humanos , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Paclitaxel/administração & dosagem , Paclitaxel/efeitos adversos , Paclitaxel/farmacocinética , Suramina/administração & dosagem , Suramina/efeitos adversos , Suramina/farmacocinética
2.
AAPS PharmSci ; 3(2): E15, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11741266

RESUMO

The penetration of paclitaxel into multilayered solid tumors is time- and concentration-dependent, a result of the drug-induced apoptosis and changes in tissue composition. This study evaluates whether this tissue penetration property applies to other highly protein-bound drugs capable of inducing apoptosis. The penetration of doxorubicin was studied in histocultures of prostate xenograft tumors and tumor specimens obtained from patients who underwent radical prostatectomy. The kinetics of drug uptake and efflux in whole tumor histocultures were studied by analyzing the average tumor drug concentration using high-pressure liquid chromatography. Spatial drug distribution in tumors and the drug concentration gradient across the tumors were studied using fluorescence microscopy. The results indicate that drug penetration was limited to the periphery for 12 hours in patient tumors and to 24 hours in the more densely packed xenograft tumors. Subsequently, the rate of drug penetration to the deeper tumor tissue increased abruptly in tumors treated with higher drug concentrations capable of inducing apoptosis (i.e., = 5 microm), but not in tumors treated with lower concentrations. These findings indicate a time- and concentration-dependent penetration of doxorubicin in solid tumors, similar to that of paclitaxel. We conclude that doxorubicin penetration in solid tumors is time- and concentration-dependent and is enhanced by drug-induced cell death.


Assuntos
Antineoplásicos/metabolismo , Doxorrubicina/metabolismo , Neoplasias da Próstata/metabolismo , Animais , Apoptose , Contagem de Células , Cromatografia Líquida de Alta Pressão , Técnicas de Cultura , Relação Dose-Resposta a Droga , Humanos , Masculino , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Transplante de Neoplasias , Fatores de Tempo
3.
J Urol ; 166(6): 2500-5, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11696818

RESUMO

PURPOSE: We previously showed that mitomycin C activity in human bladder tumors is inversely related to tumor stage and muscle invasive tumors are less sensitive than superficial tumors. Because DT-diaphorase and reduced nicotinamide adenine dinucleotide phosphate:cytochrome P450 oxidoreductase (P450R) have a role in mitomycin C bioactivation, we investigated the distribution of these enzymes as a function of depth in the bladder wall and in human bladder tumors located at different parts of the bladder. MATERIALS AND METHODS: Gene expression of DT-diaphorase and P450R relative to the expression of beta-actin was measured by reverse transcriptase-polymerase chain reaction in 4 dog and 8 human bladder tissue specimens, and in 46 human bladder tumors. DT-diaphorase activity was measured by enzymatic assay in dog and human bladders. RESULTS: Data showed decreasing expression of DT-diaphorase and P450R from urothelium to muscle layer in the normal bladder wall with higher interindividual variation in humans than in dogs (greater than 40-fold versus approximately 3-fold). The expression of DT-diaphorase and P450R in bladder tumors correlated inversely with tumor stage (p <0.05) and was significantly higher in superficial than in muscle invasive bladder disease. DT-diaphorase and P450R expression in bladder tumors was approximately 6-fold higher than in normal bladder tissue. In normal and tumor tissues DT-diaphorase expression correlated significantly with P450R (r2 = 0.33, p <0.01), while DT-diaphorase expression correlated with its enzyme activity (r2 = 0.84, p <0.01). CONCLUSIONS: Our results indicate a higher distribution of DT-diaphorase and P450R in superficial bladder tumors and tissues. Preferential enzyme distribution in superficial tumors may be a cause of the higher efficacy of intravesical mitomycin C therapy for superficial versus muscle invasive disease.


Assuntos
NAD(P)H Desidrogenase (Quinona)/metabolismo , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Neoplasias da Bexiga Urinária/enzimologia , Bexiga Urinária/enzimologia , Animais , Cães , Humanos
4.
J Pharmacol Exp Ther ; 299(2): 426-33, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11602651

RESUMO

We recently reported that acidic and basic fibroblast growth factors (aFGF and bFGF) confer a broad-spectrum chemoresistance in solid tumors, and that inhibitors of these proteins enhanced the antitumor activity of several anticancer drugs. The present study investigated the effect of FGF inhibitors on doxorubicin activity in human prostate PC3 tumors. In in vitro studies, conditioned medium (CM) obtained from histocultures of rat MAT-LyLu lung metastases and different combinations of recombinant FGF induced a 7- to 15-fold doxorubicin resistance. Suramin had no effect on the doxorubicin activity in the absence of CM or FGF, but reversed the CM- and FGF-induced resistance by > or =90% at concentrations that had no cytotoxicity (i.e., 1-17 microM suramin). In the in vivo study, immunodeficient mice bearing well established, subcutaneous PC3 tumors (approximately 100 mg in size) were treated intravenously with doxorubicin (5 mg/kg) and suramin (10 mg/kg), administered twice weekly for 3 weeks. The suramin dose, selected to yield plasma concentration of below 50 microM, had neither antitumor activity nor toxicity. Doxorubicin alone reduced tumor growth rate by approximately 60%, reduced the density of nonapoptotic tumor cells by approximately 60%, enhanced the apoptotic cell fraction by 4-fold, and reduced the body weight by approximately 15% (p < 0.05 compared with control). Addition of suramin to doxorubicin therapy did not increase weight loss but significantly enhanced the antitumor effect, resulting in complete inhibition of tumor growth, an additional 3-fold reduction in the density of nonapoptotic tumor cells, and an additional 2-fold enhancement of the apoptotic tumor cell fraction (p < 0.05 compared with all other groups). These data indicate significant enhancement of the effectiveness of doxorubicin in prostate tumors by nontoxic and subtherapeutic doses of suramin.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Antineoplásicos/uso terapêutico , Doxorrubicina/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Suramina/uso terapêutico , Algoritmos , Animais , Meios de Cultivo Condicionados , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Fator 1 de Crescimento de Fibroblastos/antagonistas & inibidores , Fator 1 de Crescimento de Fibroblastos/farmacologia , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Neoplasias da Próstata/patologia , Células Tumorais Cultivadas
5.
Cancer Res ; 61(16): 6145-50, 2001 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-11507065

RESUMO

We recently reported that acidic (aFGF) and basic (bFGF) fibroblast growth factors confer a broad spectrum chemoresistance in solid tumors, and that suramin, an inhibitor of multiple growth factors including aFGF and bFGF, enhanced the in vitro antitumor activity of several anticancer drugs including paclitaxel (Song, S., et al., Proc. Natl. Acad. Sci. USA, 97: 8658-8663, 2000). The present study investigated in vitro and in vivo interaction between paclitaxel and suramin, using human PC3-LN cells which, upon i.v. injection into immunodeficient mice, yielded lung metastases in 100% of animals. In in vitro studies, conditioned medium (CM) obtained from histocultures of rat lung metastases induced a 3-fold resistance. The addition of suramin had no effect in the absence of CM but reversed the CM-induced resistance; calculations based on the IC(50) values indicate a complete reversal in the presence of <20 microM suramin. Analysis by the combination index method indicates a synergistic interaction between paclitaxel and suramin. In in vivo studies, animals with well-established lung metastases (at least five nodules of 1 mm in diameter) were treated i.v. with paclitaxel (15 mg/kg) and suramin (10 mg/kg) administered twice weekly for 3 weeks. Single-drug therapy with paclitaxel or suramin did not reduce body weight. Suramin alone had no antitumor activity. Paclitaxel alone reduced the tumor size by approximately 75%, reduced the density of nonapoptotic cells by approximately 70% in residual tumors, and enhanced the fraction of apoptotic cells by approximately 3-fold. The addition of suramin to paclitaxel therapy enhanced the antitumor effect, resulting in an additional 5-fold reduction of tumor size, an additional 9-fold reduction of the density of nonapoptotic cells, and an additional 30% increase in the apoptotic cell fraction. These data indicate significant enhancement of the efficacy of paclitaxel by suramin and support the use of nontoxic doses of suramin with paclitaxel in the treatment of lung cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Paclitaxel/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Suramina/farmacologia , Animais , Meios de Cultivo Condicionados , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Paclitaxel/administração & dosagem , Neoplasias da Próstata/patologia , Ratos , Suramina/administração & dosagem , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Control Release ; 74(1-3): 31-46, 2001 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-11489481

RESUMO

This presentation addresses the barriers and determinants and the importance of drug-induced apoptosis in drug transport and delivery to organs and solid tumors. In particular, we examined the roles of interstitial space, drug removal by capillaries, tissue structure and tissue composition on drug distribution. Drug transport in bladder tissues is described by the distributed model which combined monodimensional Fickian diffusion and first order removal of drug by the perfusing blood. Microscopic evaluation of the spatial drug distribution in bladder, prostate and tongue indicates heterogeneous drug distribution with large and erratic concentration gradient. In general, drug distribution favors interstitial space and vasculature, with little penetration in muscles. Drug penetration into 3-dimensional solid tumors is typically 5- to 10-fold slower than in monolayer cultures. The transport of highly protein-bound drugs such as paclitaxel and doxorubicin in a solid tumor is retarded by a high tumor cell density and enhanced by drug-induced apoptosis. Accordingly, the delivery of a highly protein-bound drug to cells in a solid tumor is affected by its apoptotic effects and is therefore determined by the drug concentration and the treatment duration, i.e. treatment schedule. Under in vitro and in vivo conditions, the delivery of highly protein-bound drugs to tumor can be enhanced by using a pretreatment that induces apoptosis and reduction in cell density, and by using treatment schedules designed to take advantage of these drug-induced changes in tumor tissue composition. In conclusion, in addition to the usual processes involved in drug transport such as distribution through vascular space, transport across microvessel walls, and diffusion through interstitial space in tumor tissue, other factors including tissue structure and composition and alteration by drug-induced apoptosis are important determinants of drug distribution in organs and solid tumors.


Assuntos
Antineoplásicos/uso terapêutico , Sistemas de Liberação de Medicamentos , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/farmacocinética , Humanos , Neoplasias/metabolismo , Distribuição Tecidual
7.
Pharm Res ; 18(7): 907-13, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11496948

RESUMO

PURPOSE: We previously reported that in patient tumors the expression of the mdrl p-glycoprotein (Pgp) resulted in a lower paclitaxel-induced inhibition of DNA precursor incorporation, but a higher apoptosis (Clin. Cancer Res. 4:2949-2955, 1998). The present study was to evaluate these findings in an experimental system where the Pgp effect can be studied without confounding factors such as the intra- and inter-tumor heterogeneity associated with patient tumors. METHODS: To separate the effect of Pgp on intracellular paclitaxel accumulation from its effects on drug sensitivity, we compared the drug activity at various extracellular and intracellular drug concentrations using the human breast MCF7 tumor cells and its mdr1-transfected variant BC19 cells. RESULTS: Compared to MCF7 cells, BC19 cells showed a 9-fold higher Pgp level and >13-fold higher mdrl expression. Intracellular paclitaxel accumulation was 80-130% lower in BC19 cells when the extracellular concentrations were < or = 100 nM, but the difference was reduced to <15% differences at higher extracellular concentrations of > or = 1,000 nM. For the G2/M block effect MCF7 cells were 43-fold more sensitive than BC19 cells at equal extracellular concentration, and 3.5-fold more sensitive at comparable intracellular concentrations. On the contrary. BC19 cells were more sensitive to the apoptotic effect: BC19 cells showed equal or higher apoptosis compared to MCF7 cells at extracellular concentrations above 100 nM, and a 30-100% higher apoptosis at comparable intracellular concentrations. CONCLUSIONS: These results confirm our previous observations in patient tumors and indicate that enhanced Pgp expression is associated with enhanced sensitivity to the apoptotic effect of paclitaxel and reduced sensitivity to its G2/M block effect, via yet-unknown mechanisms that are unrelated to the effect of Pgp on intracellular drug accumulation.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Antineoplásicos Fitogênicos/farmacologia , Apoptose/genética , Paclitaxel/farmacologia , Transfecção , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/fisiologia , Apoptose/efeitos dos fármacos , Transporte Biológico Ativo/efeitos dos fármacos , Transporte Biológico Ativo/genética , Humanos , Paclitaxel/metabolismo , Transfecção/métodos , Células Tumorais Cultivadas/metabolismo
8.
J Pharmacol Exp Ther ; 298(3): 1236-42, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11504826

RESUMO

Paclitaxel is a substrate of the mdr1 P-glycoprotein (Pgp). The objective of the present study was to determine the kinetics of the Pgp-mediated efflux and its contribution to the overall efflux of paclitaxel at the clinically achievable concentration range of 1 to 1500 nM. Human breast carcinoma BC19 cells that were derived from MCF7 cells by mdr1 transfection and show a >10-fold higher level of the Pgp protein were used to measure the uptake and efflux of [(3)H]paclitaxel. A computational model of intracellular paclitaxel pharmacokinetics was developed to analyze for the Pgp efflux parameters. The results show a saturable Pgp-mediated efflux in BC19 cells; the dissociation constant was 14 nM, and the maximal efflux rate was 2.8 x 10(-4) pmol/h/cell. The contribution of Pgp-mediated efflux to the total efflux decreased with increasing extracellular drug concentrations; the Pgp efflux accounted for 86 and 34% of total efflux at 1 and 1500 nM, respectively. The validity of the model was confirmed by the close agreement between the model-predicted data and the experimentally obtained data (approximately 6% deviation) describing the effect of cell density and intracellular-to-extracellular concentration gradient on the kinetics of drug accumulation and efflux. In conclusion, our results indicate that the Pgp-mediated efflux represents a major efflux mechanism of paclitaxel at the low end of the clinically observed drug concentration range, but accounts for only a minor part of the efflux at higher concentrations in BC19 cells.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/fisiologia , Antineoplásicos Fitogênicos/farmacocinética , Paclitaxel/farmacocinética , Algoritmos , Neoplasias da Mama/metabolismo , Feminino , Genes MDR/genética , Humanos , Modelos Biológicos , Reprodutibilidade dos Testes , Transfecção , Células Tumorais Cultivadas
9.
Pharm Res ; 18(4): 488-93, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11451036

RESUMO

PURPOSE: Telomerase is a ribonucleoprotein that extends telomeres at the ends of chromosome. Increased telomerase activity is associated with cellular immortality. The currently available assay for telomerase, i.e., telomeric repeat amplification protocol (TRAP), consists of 2 steps: (a) telomerase-mediated extension of an oligonucleotide primer by the enzyme-containing extracts of cells and tissues, and (b) amplification of the telomerase-extended primer products by polymerase chain reaction (PCR) and detection of the PCR products. It is generally accepted that the current TRAP assay lacks quantitative precision. The present study was to develop a quantitative telomerase assay with greater precision and sensitivity. METHODS: This new method used the primer extension method as in TRAP, plus the following modifications: (a) used a lysis buffer that yielded complete lysis of nuclei; (b) removal of PCR inhibitors by phenol/chloroform extraction after primer extension; and (c) used primers for the internal standard that were designed to reduce their competition with the telomerase products for PCR. RESULTS: The modified method showed a good correlation (r2 = 0.99, P < 0.001) between telomerase amount (expressed as total protein in cell lysate) and its activity (expressed as telomerase products). Compared to the conventional TRAP, the new method (a) was more sensitive (average of 5.5-fold in cultured cancer cells and >5.9-fold in patient tumors), (b) had a lower inter- and intra-day variability (>3fold), and (c) showed a 2 to 4-fold broader range of linearity in the standard curve. The higher assay sensitivity further enabled the use of a nonradioactive method, i.e., ethidium bromide staining of DNA, to detect the TRAP products, as opposed to the use of radioactive nucleotide and the more labor-intensive autoradiography mandated by the conventional TRAP. CONCLUSION: We report here a quantitative assay for telomerase activity in cultured human cancer cells and patient tumors.


Assuntos
Telomerase/metabolismo , Bioensaio/métodos , Humanos , Reação em Cadeia da Polimerase/métodos , Células Tumorais Cultivadas/enzimologia
10.
Invest New Drugs ; 19(2): 113-23, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11392446

RESUMO

This report addresses the determinants of the rate and extent of paclitaxel accumulation in tumors. In a 2-dimensional system such as monolayers where the drug is directly in contact with tumor cells, drug accumulation is determined by the extracellular-to-intracellular concentration gradient, the drug binding to extracellular and intracellular macromolecules, the presence of the mdrl p-glycoprotein (Pgp). and the time-dependent and drug concentration-dependent changes in tubulins and cell density. Intracellular pharmacokinetic models were developed to depict the effects of these parameters. Computer simulation results indicate that at the clinically relevant concentration range of 1 to 1,000 nM, (a) the binding affinity and the number of intracellular saturable drug binding sites are important for drug accumulation at low and high extracellular concentrations, respectively, (b) saturation in the drug binding to the high affinity intracellular binding sites (e.g., tubulin/microtubule) occurs at extracellular drug concentration above 100 nM, (c) treatment with 1,000 nM paclitaxel for >4 hr results in increased levels of tubulin/microtubule and consequently increased intracellular drug accumulation, whereas the continued cell proliferation after treatment with low drug concentrations results in reduced intracellular accumulation, and (d) saturation of Pgp in mdr1-transfected cells occurs at the high end of the clinically relevant concentration range. In a 3-dimensional system such as the solid tumor histocultures, which contain tumor cells as well as stromal cells, the drug accumulation into the inner cell layers is determined by the unique properties of solid tumors, including tumor cell density and spatial arrangement of tumor and stromal tissues. Most interestingly, drug penetration is modulated by the drug-induced apoptosis; the reduced cell density due to apoptosis results in an enhancement of the rate of drug penetration into the inner cell layers of solid tumors. In conclusion, the uptake, accumulation, and retention of paclitaxel in solid tumors are determined by (a) factors that are independent of biological changes in tumor cells induced by paclitaxel, i.e., ratio of extracellular and intracellular concentrations, and drug binding to extracellular and intracellular macromolecules, and (b) factors that are dependent on the time- and drug concentration-dependent biological changes induced by paclitaxel, i.e., induction of apoptosis, enhancement of tubulin/microtubule production, and induction of Pgp expression.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Antineoplásicos Fitogênicos/farmacocinética , Neoplasias/metabolismo , Paclitaxel/farmacocinética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Apoptose/fisiologia , Sítios de Ligação/fisiologia , Membrana Celular/metabolismo , Difusão , Humanos , Tubulina (Proteína)/metabolismo , Células Tumorais Cultivadas
11.
Clin Cancer Res ; 7(5): 1313-9, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11350900

RESUMO

Mitomycin C (MMC) is activated by DT-diaphorase (DTD) and cytochrome P450 reductase (P450R). In cancer cell lines, MMC cytotoxicity is correlated with DTD and P450R expression levels. The present study investigated the relationship between enzyme expression/activity and MMC cytotoxicity in patient bladder tumors. DTD and P450R expression was detected by competitive reverse transcription-PCR and their activity was measured by bioreductive assays. The expression of DTD and P450R in patient tumors (n = 29), as ratios to beta-actin levels, varied from 0 to 90% and 0 to 29%, respectively. The DTD expression was significantly correlated with P450R expression (r(2), 0.32; P < 0.01), whereas the average DTD level was 2-fold higher than that of P450R (P < 0.01). Among the 29 tumors, 21 provided sufficient materials to evaluate tumor sensitivity to MMC. The concentration of MMC required to produce 50% inhibition (IC(50)) of DNA precursor incorporation for a 2-h treatment ranged from 0.17 to 18.1 microg/ml, indicating a 110-fold intertumor variation, with the high-grade and more invasive tumors being less chemosensitive compared with the low-grade and less invasive tumors. Tumor sensitivity to MMC, as indicated by the inverse of IC(50) values, was positively correlated with the expression of DTD (r(2), 0.28; P < 0.05) and P450R (r(2), 0.26; P < 0.05). Multivariate analysis indicates DTD expression and P450R expression as better determinants of MMC activity compared with other pathobiological factors (e.g., tumor grade, stage, and labeling index) that have been shown to significantly correlate with MMC activity. Eleven tumors were studied for the relationship between gene expression level and enzyme activity of DTD and P450R. The DTD activity was significantly correlated with the gene expression level (r(2), 0.84; P < 0.001). For P450R, there is a trend of a correlation between enzyme activity and its mRNA level, but the correlation was not statistically significant (r(2), 0.28; P = 0.09). These data indicate that the sensitivity of patient bladder tumors to MMC is correlated with the expression of DTD and P450R in tumors and suggest that the lower expression of these enzymes in the high-grade and more invasive tumors is a cause of the lower efficacy of intravesical MMC in these tumors.


Assuntos
Antibióticos Antineoplásicos/farmacologia , DNA de Neoplasias/efeitos dos fármacos , Mitomicina/farmacologia , NAD(P)H Desidrogenase (Quinona)/biossíntese , NADPH-Ferri-Hemoproteína Redutase/biossíntese , Neoplasias da Bexiga Urinária/enzimologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , DNA de Neoplasias/biossíntese , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Expressão Gênica , Humanos , Concentração Inibidora 50 , Masculino , Pessoa de Meia-Idade , NAD(P)H Desidrogenase (Quinona)/genética , NADPH-Ferri-Hemoproteína Redutase/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias da Bexiga Urinária/genética
12.
J Natl Cancer Inst ; 93(8): 597-604, 2001 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-11309436

RESUMO

BACKGROUND: Intravesical chemotherapy (i.e., placement of the drug directly in the bladder) with mitomycin C is beneficial for patients with superficial bladder cancer who are at high risk of recurrence, but standard therapy is empirically based and patient response rates have been variable, in part because of inadequate drug delivery. We carried out a prospective, two-arm, randomized, multi-institutional phase III trial to test whether enhancing the drug's concentration in urine would improve its efficacy. METHODS: Patients with histologically proven transitional cell carcinoma and at high risk for recurrence were eligible for the trial. Patients in the optimized-treatment arm (n = 119) received a 40-mg dose of mitomycin C, pharmacokinetic manipulations to increase drug concentration by decreasing urine volume, and urine alkalinization to stabilize the drug. Patients in the standard-treatment arm (n = 111) received a 20-mg dose without pharmacokinetic manipulations or urine alkalinization. Both treatments were given weekly for 6 weeks. Primary endpoints were recurrence and time to recurrence. Treatment outcome was examined by use of Kaplan-Meier analysis with log-rank tests. Statistical tests were two-sided. RESULTS: Patients in the two arms did not differ in demographics or history of intravesical therapy. Dysuria occurred more frequently in the optimized arm but did not lead to more frequent treatment termination. In an intent-to-treat analysis, patients in the optimized arm showed a longer median time to recurrence (29.1 months; 95% confidence interval [CI] = 14.0 to 44.2 months) and a greater recurrence-free fraction (41.0%; 95% CI = 30.9% to 51.1%) at 5 years than patients in the standard arm (11.8 months; 95% CI = 7.2 to 16.4 months) and 24.6% (95% CI = 14.9% to 34.3%) (P =.005, log-rank test for time to recurrence). Improvements were found in all risk groups defined by tumor stage, grade, focality, and recurrence. CONCLUSIONS: This study identified a pharmacologically optimized intravesical mitomycin C treatment with statistically significantly enhanced efficacy.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Carcinoma de Células de Transição/tratamento farmacológico , Mitomicina/administração & dosagem , Neoplasias da Bexiga Urinária/tratamento farmacológico , Administração Intravesical , Adulto , Idoso , Idoso de 80 Anos ou mais , Antibióticos Antineoplásicos/efeitos adversos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mitomicina/efeitos adversos , Recidiva Local de Neoplasia/prevenção & controle , Estudos Prospectivos , Fatores de Risco
13.
Cancer Invest ; 19(2): 127-36, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11296617

RESUMO

From February 1993 through July 1994, 37 patients with stage III-IV squamous cell carcinomas of the oral cavity, oropharynx, or hypopharynx (stage II-IV) were registered to a treatment regimen consisting of preoperative continuous infusion cisplatin (80 mg/m2/80 hours) with hyperfractionated external beam radiotherapy (9.1 Gy/7 fractions of 1.3 Gy BID), surgical resection, intraoperative radiotherapy (7.5 Gy), and postoperative radiotherapy (40 Gy) with concurrent cisplatin (100 mg/m2 x 2 courses). The objectives of the regimen were to improve patient compliance while also increasing treatment intensity. The purpose of this article is to report the local, regional (nodal), and distant disease control of these patients after an extended time at risk (median 40 months). Overall compliance (73%), local control at primary site (97%), and regional nodal control (95%) were excellent. The rate of distant metastasis was 19%. Absolute survival at 48 months was 45.9%.


Assuntos
Carcinoma de Células Escamosas/cirurgia , Cisplatino/uso terapêutico , Neoplasias de Cabeça e Pescoço/cirurgia , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/radioterapia , Quimioterapia Adjuvante/efeitos adversos , Terapia Combinada/efeitos adversos , Fracionamento da Dose de Radiação , Seguimentos , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/mortalidade , Neoplasias de Cabeça e Pescoço/radioterapia , Humanos , Neoplasias Hipofaríngeas/tratamento farmacológico , Neoplasias Hipofaríngeas/radioterapia , Neoplasias Hipofaríngeas/cirurgia , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/radioterapia , Neoplasias Bucais/cirurgia , Metástase Neoplásica , Estadiamento de Neoplasias , Neoplasias Orofaríngeas/tratamento farmacológico , Neoplasias Orofaríngeas/radioterapia , Neoplasias Orofaríngeas/cirurgia , Cooperação do Paciente , Dosagem Radioterapêutica , Taxa de Sobrevida , Fatores de Tempo , Resultado do Tratamento
14.
J Pharmacol Exp Ther ; 296(3): 1035-42, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11181938

RESUMO

The limited penetration of paclitaxel into solid tumors may limit its therapeutic efficacy. We recently showed a correlation between an increase in interstitial space and an enhancement of drug delivery in solid tumors. The present study evaluated whether this observation can be used to develop a treatment strategy, where an apoptosis-inducing pretreatment with paclitaxel is used to enhance its own delivery to solid tumors. In histocultures of human pharynx FaDu xenograft tumors, pretreatment with 1 microM nonradiolabeled paclitaxel, which resulted in approximately 25% apoptosis and a 25% reduction in cell density, enhanced the penetration rate of [(3)H]paclitaxel. Likewise, dividing a total drug exposure to two treatments, separated by an interval to allow apoptosis to occur, resulted in higher drug penetration rate and accumulation compared with giving the same drug exposure continuously. Similar results were obtained in rats bearing subcutaneously implanted prostate MAT-LyLu tumors; fractionation of the dose, to include 1) a pretreatment that yielded sufficient and clinically relevant plasma concentration to induce apoptosis and 2) a second dose given at an interval selected to allow apoptosis and reduction in tumor cell density to occur, resulted in higher tumor concentration compared with other treatments using the same total dose but either did not include an apoptosis-inducing pretreatment or did not allow for apoptosis to occur. We conclude that the pharmacological effect of paclitaxel affects its own delivery to solid tumors and that modifications of the paclitaxel treatment schedule can enhance drug delivery in solid tumors.


Assuntos
Antineoplásicos Fitogênicos/farmacocinética , Apoptose , Neoplasias Experimentais/tratamento farmacológico , Paclitaxel/farmacocinética , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/uso terapêutico , Contagem de Células , Modelos Animais de Doenças , Esquema de Medicação , Interações Medicamentosas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/metabolismo , Paclitaxel/administração & dosagem , Paclitaxel/uso terapêutico , Trítio , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Pharm Res ; 18(12): 1655-9, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11785683

RESUMO

PURPOSE: Telomeres are specific DNA structure at the ends of chromosomes to protect chromosomes from fusion, recombination, and degradation. Telomere length changes are implicated in cell senescence, aging, tumorigenesis, and DNA repair. The standard method for measuring telomere length is Southern blot analysis. This method has several disadvantages, i.e., loss of DNA during membrane blotting, high background due to nonspecific binding of telomere probe to membrane, and loss of telomeric signal due to extensive washing. These limitations resulted in a low signal-to-noise ratio and, therefore, reduced sensitivity and reproducibility. The multi-step Southern blot is also highly labor-intensive. The present study was to develop a more quantitative assay of telomeric amount and length (TALA). METHODS: TALA was based on solution hybridization and did not require blotting, prehybridization, and washing. The major steps were (a) DNA preparation and digestion with restriction endonucleases, (b) hybridization between DNA and telomeric probe, (c) agarose gel electrophoresis, and (d) autoradiography and data analysis. RESULTS: The telomere amount measured by TALA was linearly correlated with the amount of DNA analyzed (r2 = 0.985, P < 0.01). The telomere length measured by TALA also correlated with the telomere length determined by fluorescence in situ hybridization (r2 = 0.99, P < 0.01). Compared to the Southern blot analysis, TALA showed a 4-fold greater sensitivity, 4.6-fold higher signal-to-noise ratio, >2 fold-higher reproducibility, and 4-fold less time requirement. CONCLUSION: We report here a rapid, sensitive, and quantitative assay for measuring telomere length and amount.


Assuntos
DNA/química , Telômero/química , Hibridização In Situ , Telômero/ultraestrutura , Fatores de Tempo , Células Tumorais Cultivadas
16.
Proc Natl Acad Sci U S A ; 97(15): 8658-63, 2000 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-10890892

RESUMO

Based on the observation that removal of tumors from metastatic organs reversed their chemoresistance, we hypothesized that chemoresistance is induced by extracellular factors in tumor-bearing organs. By comparing chemosensitivity and proteins in different tumors (primary vs. metastases) and different culture systems (tumor fragment histocultures vs. monolayer cultures derived from the same tumor), we found elevated levels of acidic (aFGF) and basic (bFGF) fibroblast growth factors in the conditioned medium (CM) of solid and metastatic tumors. These CM induced broad spectrum resistance to drugs with diverse structures and action mechanisms (paclitaxel, doxorubicin, 5-fluorouracil). Inhibition of bFGF by mAb and its removal by immunoprecipitation resulted in complete reversal of the CM-induced chemoresistance, whereas inhibition/removal of aFGF resulted in partial reversal. Using CM that had been depleted of aFGF and/or bFGF and subsequently reconstituted with respective human recombinant proteins, we found that bFGF but not aFGF induced chemoresistance whereas aFGF amplified the bFGF effect. aFGF and bFGF fully accounted for the CM effect, indicating these proteins as the underlying mechanism of the chemoresistance. The FGF-induced resistance was not due to reduced intracellular drug accumulation or altered cell proliferation. We further showed that an inhibitor of aFGF/bFGF (suramin) enhanced the in vitro and in vivo activity of chemotherapy, resulting in shrinkage and eradication of well established human lung metastases in mice without enhancing toxicity. These results indicate elevated levels of extracellular aFGF/bFGF as an epigenetic mechanism by which cancer cells elude cytotoxic insult by chemotherapy, and provide a basis for designing new treatment strategies.


Assuntos
Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Fator 1 de Crescimento de Fibroblastos/fisiologia , Fator 2 de Crescimento de Fibroblastos/fisiologia , Fluoruracila/farmacologia , Paclitaxel/farmacologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Animais , Divisão Celular/efeitos dos fármacos , Meios de Cultivo Condicionados , Resistencia a Medicamentos Antineoplásicos/fisiologia , Fator 1 de Crescimento de Fibroblastos/antagonistas & inibidores , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Metástase Linfática/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Ratos , Suramina/farmacologia , Células Tumorais Cultivadas
17.
J Pharmacol Exp Ther ; 293(3): 761-70, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10869374

RESUMO

The intracellular pharmacokinetics of paclitaxel is closely related to its pharmacodynamics. Although drug transport across the cell membrane and extracellular and intracellular drug binding have been shown to affect intracellular drug accumulation, their quantitative relationship is unknown. This study was designed to establish a mathematical model for computing the intracellular paclitaxel pharmacokinetics. As a starting point, the model assumes drug transport into and out of cells via passive diffusion. Experimental data on the intracellular pharmacokinetics of [(3)H]paclitaxel were obtained using monolayer cultures of human breast MCF7 tumor cells, which have negligible expression of the mdr1 P-glycoprotein. The results indicate that, in addition to drug binding and microtubule concentration, changes in cell number due to cell growth and drug effects also affected intracellular drug accumulation. A kinetic model was developed to describe several concomitant processes: 1) saturable drug binding to extracellular proteins, 2) saturable and nonsaturable drug binding to intracellular components, 3) time- and concentration-dependent drug depletion from culture medium, 4) cell density-dependent drug accumulation, and 5) time- and drug concentration-dependent enhancement of tubulin concentration. The model was validated by the close prediction (<7% deviation) of the effects of extracellular-to-intracellular concentration gradient and cell density on the kinetics of drug accumulation and efflux. This model was used to predict the effects of changing several parameters (number and binding affinity of intracellular binding sites, free fraction, and concentration of drug in extracellular fluid) on intracellular drug accumulation. In conclusion, the computational model of intracellular paclitaxel pharmacokinetics provides the means to predict drug concentration in cells.


Assuntos
Antineoplásicos Fitogênicos/farmacocinética , Paclitaxel/farmacocinética , Contagem de Células , Humanos , Matemática , Microtúbulos/efeitos dos fármacos , Modelos Biológicos , Paclitaxel/farmacologia , Tubulina (Proteína)/metabolismo , Células Tumorais Cultivadas
18.
Cancer Chemother Pharmacol ; 45(4): 265-72, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10755313

RESUMO

PURPOSES: The pharmacodynamics of most drugs follow the empirical relationship, C(n) x T = h, where C is drug concentration, T is exposure time and h is drug exposure constant. The value of n indicates the relative importance of C and T in determining the effect. An n value greater than 1.0 indicates that for two infusions that produce the same C x T, a short infusion that delivers high concentrations over a short duration will produce a greater C(n) x T and therefore a greater effect, compared to a long infusion that delivers lower concentrations. The reverse is true for an n value less than 1.0 and would support the use of a slow infusion. Hence, it is important to determine the n values and whether the n value significantly differs from 1.0. This report describes a three-step method for this purpose. METHODS: First, we obtained experimental data on the relationship between drug concentration, treatment time and effect, and analyzed the data with a three-dimensional surface response method to obtain the pharmacodynamic model parameters and the magnitude of data variability. The experiments used mitomycin C and two human cancer cell lines, i.e. bladder RT4 and pharynx FaDu cells. The n values obtained from four experiments ranged from 1.04 to 1.16 for FaDu cells and from 1.14 to 1.46 for RT4 cells. The variability in the effect data decreased from 11.9% at 0% effect to 6.14% at 100% effect. Second, these results were used with Monte Carlo simulations to generate 100 concentration-time-effect data sets, which contained randomly and normally distributed data variability comparable to the experimentally observed variability, for each experimentally determined n value. This is analogous to performing 100 experiments under the same experimental conditions. Third, we analyzed the simulated data sets to obtain 100 estimated n values. The frequency with which these estimated n values fell above or below 1.0 indicated the probability that the experimentally determined n value used in the Monte Carlo simulations was truly different from 1.0. We defined this frequency for individual experiments as F(one), and calculated the overall probability for multiple experiments (F(multiple)). A probability of greater than 97.5% (i.e. P < 0.05 for a two-tailed test) was considered statistically significant. RESULTS: Analysis of the mitomycin C pharmacodynamic data yielded F(one) and F(multiple) of 99% to 100% for FaDu and RT4 cells, indicating that the n values for these cells were significantly higher than 1.0. A comparison of the statistical significance of the n value analyzed by the three-step pharmacodynamic analysis method, a conventional statistical method such as the Student's t-test and nonlinear regression analysis, indicated two advantages for the pharmacodynamic method: fewer experiments were required (theoretically only one experiment with three replicates would be sufficient) and a higher statistical significance of the n value was obtained. CONCLUSIONS: In summary, the three-step pharmacodynamic study design and analysis method can be used to define the relative importance of drug concentration and treatment time on drug effect.


Assuntos
Antineoplásicos/farmacologia , Algoritmos , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/farmacologia , Antibióticos Antineoplásicos/uso terapêutico , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Humanos , Mitomicina/farmacocinética , Mitomicina/farmacologia , Mitomicina/uso terapêutico , Modelos Biológicos , Método de Monte Carlo , Análise de Regressão , Fatores de Tempo , Células Tumorais Cultivadas
19.
Arch Otolaryngol Head Neck Surg ; 125(12): 1313-8, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10604408

RESUMO

OBJECTIVE: To determine the feasibility, toxicity, and compliance of an intense treatment regimen for patients with advanced, previously untreated, resectable head and neck squamous cell carcinomas. DESIGN: Prospective, nonrandomized, controlled (phase 1 or 2) clinical trial; median time at risk, 25 months (range, 7 days to 36 months). SETTING: Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus. PATIENTS: Forty-three patients (median age, 59 years; range, 32-76 years) with resectable, previously untreated stage III or IV squamous cell carcinomas of the oral cavity, oropharynx, or hypopharynx or stage II squamous cell carcinomas of the hypopharynx (referred sample of patients). INTERVENTIONS: Days 1 to 4, perioperative, slightly accelerated, hyperfractionated radiotherapy (9.1 Gy) to off cord fields; days 1 to 3, cisplatin, 30 mg/m2 per day; day 4, surgical resection and intraoperative radiotherapy boost (7.5 Gy); days 45 to 52, postoperative radiotherapy (40 Gy to the primary site and upper neck and 45 Gy to the supraclavicular areas); days 24, 45, and 66, paclitaxel, 135 mg/m2 per 24 hours, with routine granulocyte colony-stimulating factor support; and days 25 and 46, cisplatin, 100 mg/m2. MAIN OUTCOME MEASURES: Toxicity, compliance, local control, and distant metastatic rates. RESULTS: Patient compliance was 91% (39 of 43 patients), but protocol compliance was only 58% (25 of 43 patients), reflecting increased toxicity of the systemic regimen (2 [5%] of the 43 patients experienced grade 5 hematologic toxicity due to the regimen; 16 [37%], grade 4; and 10 [23%], grade 3). Local-regional control was 92% (23 of 25 patients), and the distant metastatic rate was 8% (2 of 25) in patients completing treatment per protocol. One patient had surgical salvage of a second primary tumor. CONCLUSIONS: Local control and patient compliance were encouraging, but systemic toxicity was unacceptable. Thus, the paclitaxel was changed to a weekly regimen.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/radioterapia , Cisplatino/uso terapêutico , Neoplasias de Cabeça e Pescoço/radioterapia , Paclitaxel/uso terapêutico , Adulto , Idoso , Antineoplásicos/efeitos adversos , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/cirurgia , Cisplatino/efeitos adversos , Terapia Combinada , Relação Dose-Resposta à Radiação , Estudos de Viabilidade , Feminino , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/cirurgia , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Paclitaxel/efeitos adversos , Cooperação do Paciente , Taxa de Sobrevida
20.
Cancer Chemother Pharmacol ; 44(3): 241-8, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10453726

RESUMO

PURPOSE: We have previously shown that paclitaxel, when dissolved in water and instilled into the bladder, readily penetrates the urothelium. The FDA-approved formulation uses Cremophor and ethanol to dissolve paclitaxel. In the present study, the effects of this solvent system on the urine, bladder tissue, and plasma pharmacokinetics of intravesical paclitaxel were evaluated. METHODS: Plasma, urine, and tissue pharmacokinetics were determined in five dogs treated for 120 min with paclitaxel (500 microg per 20 ml of 0.22% w/v Cremophor and 0.21% v/v ethanol) by intravesical instillation. Equilibrium dialysis was used to determine the free fraction of paclitaxel and the presence of Cremophor micelles was verified using a fluorescent probe method. RESULTS: The average bladder tissue concentration was > 1600-fold higher than the plasma concentration. Comparison of the results for paclitaxel dissolved in Cremophor/ethanol with our previous results of paclitaxel dissolved in water (500 microg per 20 ml) indicates that Cremophor/ethanol decreased the paclitaxel partition across the urothelium and reduced the average bladder tissue concentration by 75%, but did not alter the rate of paclitaxel penetration across the bladder wall, the urine pharmacokinetics or the plasma pharmacokinetics of paclitaxel. For Cremophor, the urine concentrations during the 120-min treatment ranged from 0.12% to 0.22%, and the concentration in bladder tissue from 0.00004% to 0.0009%. The threshold Cremophor concentration for micelle formation was 0.008%. We found that ethanol at concentrations up to 1% and Cremophor at concentrations below 0.01% did not alter the free fraction of paclitaxel, whereas Cremophor at higher concentrations, i.e. 0.065% and 0.25%, significantly reduced the free fraction by two- to six-fold, respectively. These results indicate that during intravesical instillation of the FDA-approved paclitaxel formulation, the concentration of Cremophor in urine was sufficient to form micelles, resulting in sequestration of paclitaxel into micelles, reduction in the free fraction of paclitaxel and consequently a reduction in paclitaxel penetration across the urothelium. In contrast, the Cremophor concentrations in bladder tissue were inadequate to form micelles and thus did not alter the drug penetration through the bladder tissue. CONCLUSIONS: We conclude that intravesical paclitaxel treatment using the FDA-approved formulation provides a significant bladder tissue targeting advantage, although the advantage is lower than when paclitaxel is dissolved in water.


Assuntos
Antineoplásicos Fitogênicos/farmacocinética , Paclitaxel/farmacocinética , Polietilenoglicóis/administração & dosagem , Bexiga Urinária/metabolismo , Animais , Antineoplásicos Fitogênicos/sangue , Antineoplásicos Fitogênicos/urina , Cães , Feminino , Masculino , Modelos Biológicos , Paclitaxel/sangue , Paclitaxel/urina , Veículos Farmacêuticos , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...