Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Br J Cancer ; 111(4): 716-25, 2014 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-24921920

RESUMEN

BACKGROUND: A frequent mechanism of acquired multidrug resistance in human cancers is overexpression of ATP-binding cassette transporters such as the Multi-Drug Resistance Protein 1 (MDR-1). Nutlin-3, an MDM2-p53 antagonist, has previously been reported to be a competitive MDR-1 inhibitor. METHODS: This study assessed whether the structurally diverse MDM2-p53 antagonists, MI-63, NDD0005, and RG7388 are also able to modulate MDR-1 function, particularly in p53 mutant neuroblastoma cells, using XTT-based cell viability assays, western blotting, and liquid chromatography-mass spectrometry analysis. RESULTS: Verapamil and the MDM2-p53 antagonists potentiated vincristine-mediated growth inhibition in a concentration-dependent manner when used in combination with high MDR-1-expressing p53 mutant neuroblastoma cell lines at concentrations that did not affect the viability of cells when given alone. Liquid chromatography-mass spectrometry analyses showed that verapamil, Nutlin-3, MI-63 and NDD0005, but not RG7388, led to increased intracellular levels of vincristine in high MDR-1-expressing cell lines. CONCLUSIONS: These results show that in addition to Nutlin-3, other structurally unrelated MDM2-p53 antagonists can also act as MDR-1 inhibitors and reverse MDR-1-mediated multidrug resistance in neuroblastoma cell lines in a p53-independent manner. These findings are important for future clinical trial design with MDM2-p53 antagonists when used in combination with agents that are MDR-1 substrates.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Neuroblastoma/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP , Antineoplásicos/metabolismo , Línea Celular Tumoral , Cisplatino/farmacología , Doxorrubicina/farmacología , Sinergismo Farmacológico , Humanos , Imidazoles/farmacología , Indoles/farmacología , Concentración 50 Inhibidora , Neuroblastoma/metabolismo , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Pirrolidinas/farmacología , Compuestos de Espiro/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Verapamilo/farmacología , Vincristina/metabolismo , Vincristina/farmacología , para-Aminobenzoatos/farmacología
2.
J Clin Oncol ; 41(28): 4453-4454, 2023 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-37757592

RESUMEN

A dosage formula has been derived from a retrospective analysis of carboplatin pharmacokinetics in 18 patients with pretreatment glomerular filtration rates (GFR) in the range of 33 to 136 mL/min. Carboplatin plasma clearance was linearly related to GFR (r = 0.85, P less than .00001) and rearrangements of the equation describing the correlation gave the dosage formula dose (mg) = target area under the free carboplatin plasma concentration versus time curve (AUC) x (1.2 x GFR + 20). In a prospective clinical and pharmacokinetic study the formula was used to determine the dose required to treat 31 patients (GFR range, 33 to 135 mL/min) with 40 courses of carboplatin. The target AUC was escalated from 3 to 8 mg carboplatin/mL/min. Over this AUC range the formula accurately predicted the observed AUC (observed/predicted ratio 1.24 +/- 0.11, r = 0.886) and using these additional data, the formula was refined. Dose (mg) = target AUC x (GFR + 25) is now the recommended formula. AUC values of 4 to 6 and 6 to 8 mg/mL. min gave rise to manageable hematological toxicity in previously treated and untreated patients, respectively, and hence target AUC values of 5 and 7 mg/mL min are recommended for single-agent carboplatin in these patient groups. Pharmacokinetic modeling demonstrated that the formula was reasonably accurate regardless of whether a one- or two-compartment model most accurately described carboplatin pharmacokinetics, assuming that body size did not influence nonrenal clearance. The validity of this assumption was demonstrated in 13 patients where no correlation between surface area and nonrenal clearance was found (r = .31, P = .30). Therefore, the formula provides a simple and consistent method of determining carboplatin dose in adults. Since the measure of carboplatin exposure in the formula is AUC, and not toxicity, it will not be influenced by previous or concurrent myelosuppressive therapy or supportive measures. The formula is therefore applicable to combination and high-dose studies as well as conventional single-agent therapy, although the target AUC for carboplatin will need to be redefined for combination chemotherapy.

3.
Br J Cancer ; 106(8): 1386-94, 2012 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-22415236

RESUMEN

BACKGROUND: Combined targeting of MAPK and PI3K signalling pathways may be necessary for optimal therapeutic activity in cancer. This study evaluated the MEK inhibitors AZD6244 and PD0325901, alone and in combination with the dual mTOR/PI3K inhibitor NVP-BEZ235 or the PI3K inhibitor GDC-0941, in three colorectal cancer cell lines. METHODS: Growth inhibition, survival and signal transduction were measured using the Sulforhodamine B assay, clonogenicity and western blotting, respectively, in HCT116, HT29 and DLD1 cell lines. RESULTS: All MEK/PI3K inhibitor combinations exhibited marked synergistic growth inhibition; however, GDC-0941 displayed greater synergy in combination with either MEK inhibitor. NVP-BEZ235 exhibited stronger inhibition of 4EBP1 phosphorylation, and similar inhibition of S6 and AKT phosphorylation, compared with GDC-0941. Both PD0325901 and AZD6244 inhibited ERK phosphorylation, and with MEK/PI3K inhibitor combinations inhibition of S6 phosphorylation was increased. The reduced synergy exhibited by NVP-BEZ235 in combination with MEK inhibitors, compared with GDC-0941, may be due to inhibition of mTOR, and the addition of the mTORC1/2 inhibitor KU0063794 compromised the synergy of GDC-0941:PD0325901 combinations. CONCLUSION: These studies confirm that dual targeting of PI3K and MEK can induce synergistic growth inhibition; however, the combination of specific PI3K inhibitors, rather than dual mTOR/PI3K inhibitors, with MEK inhibitors results in greater synergy.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Benzamidas/farmacología , Bencimidazoles/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Difenilamina/análogos & derivados , Difenilamina/farmacología , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Humanos , Imidazoles/farmacología , Indazoles/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Pirimidinas/farmacología , Quinolinas/farmacología , Transducción de Señal , Relación Estructura-Actividad , Sulfonamidas/farmacología , Serina-Treonina Quinasas TOR/metabolismo
4.
Br J Cancer ; 104(12): 1869-76, 2011 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-21610703

RESUMEN

BACKGROUND: Thiothymidine (S(4)TdR) can be incorporated into DNA and sensitise cells to DNA damage and cell death following exposure to UVA light. Studies were performed to determine if the combination of S(4)TdR and UVA could be an effective treatment for bladder cancer. METHODS: Uptake and incorporation of S(4)TdR was determined in rat and human bladder tumour cell lines. Measures of DNA crosslinking and apoptosis were also performed. In vivo activity of the combination of S(4)TdR and UVA was investigated in an orthotopic model of bladder cancer in rats. RESULTS: Thiothymidine (200 µM) replaced up to 0.63% of thymidine in rat and tumour bladder cancer cells. The combination of S(4)TdR (10-200 µM) and UVA (1-5 kJ m(-2)) caused apoptosis and cell death at doses that were not toxic alone. Addition of raltitrexed (Astra Zeneca, Alderley Edge, Cheshire, UK) increased the incorporation of S(4)TdR into DNA (up to 20-fold at IC(5)) and further sensitised cells to UVA. Cytotoxic effect was associated with crosslinking of DNA, at least partially to protein. Intravenous administration of S(4)TdR, in combination with UVA delivered directly to the bladder, resulted in an antitumour effect in three of five animals treated. CONCLUSION: These data indicate that the combination of S(4)TdR and UVA has potential as a treatment for bladder cancer, and give some insight into the mechanism of action. Further work is necessary to optimise the delivery of the two components.


Asunto(s)
Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Timidina/análogos & derivados , Terapia Ultravioleta , Neoplasias de la Vejiga Urinaria/terapia , Animales , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Daño del ADN , Femenino , Humanos , Quinazolinas/farmacología , Ratas , Ratas Endogámicas F344 , Tiofenos/farmacología , Timidina/metabolismo , Timidina/uso terapéutico , Timidina/toxicidad , Neoplasias de la Vejiga Urinaria/patología
5.
Br J Cancer ; 102(2): 342-50, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20010939

RESUMEN

BACKGROUND: Cellular proliferation, driven by cyclin-dependent kinases (CDKs) and their cyclin partners, is deregulated in cancer. Anti-estrogens, such as tamoxifen, antagonise estrogen-induced ERalpha transactivation of cyclin D1, resulting in reduced CDK4/6 activity, p27(Kip1)-mediated inhibition of CDK2 and growth arrest. We hypothesised that direct inhibition of CDK2 and CDK1 may overcome the major clinical problem of anti-estrogen resistance. METHODS: The cellular effects of CDK2/1 siRNA knockdown and purine-based CDK2/1 inhibitors, NU2058 and NU6102, were measured in anti-estrogen-sensitive and resistant breast cancer cell lines. RESULTS: CDK2 knockdown caused G1 accumulation, whereas CDK1 depletion caused G2/M slowing, and dual CDK1/2 depletion resulted in further G2/M accumulation and cell death in both anti-estrogen-sensitive and resistant cells, confirming CDK2 and CDK1 as targets for breast cancer therapy. In contrast to tamoxifen, which only affected hormone-sensitive cells, NU2058 and NU6102 reduced CDK2-mediated phosphorylation of pRb, E2F transcriptional activity and proliferation, ultimately resulting in cell death, in both anti-estrogen-sensitive and resistant cells. Both drugs caused G2/M arrest, reflective of combined CDK2/1 knockdown, with a variable degree of G1 accumulation. CONCLUSION: These studies confirm the therapeutic potential of CDK2 and CDK1 inhibitors for cancer therapy, and support their use as an alternative treatment for endocrine-resistant breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Proteína Quinasa CDC2/metabolismo , Proliferación Celular/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias de la Mama , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Moduladores de los Receptores de Estrógeno/farmacología , Femenino , Humanos
6.
Cancer Res ; 54(17): 4684-90, 1994 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-8062265

RESUMEN

ZENECA ZD0490 is a recombinant ricin A-chain-containing immunotoxin that recognizes an antigen that is expressed on approximately 65% of colorectal tumors. The antigen CA242 is recognized by a mouse monoclonal antibody designated C242. C242 antibody was conjugated to recombinant ricin A-chain via a methyl-hindred disulfide linker which confers in vivo stability. ZD0490 was extremely potent against colorectal cell lines CoLo201 and CoLo205, which express the CA242 antigen. ZD0490 activity was determined in vitro by both protein synthesis inhibition (50% inhibitory concentrations of 1-20 ng/ml after 24-h exposure) and clonogenic assay (76-95% cell kill after 24-h exposure to a 50% inhibitory concentration for protein synthesis inhibition; > 99.99% cell kill at 1000 ng/ml). This in vitro activity was translated to in vivo efficacy where single dose i.v. administration of 2.5 mg/kg of ZD0490 was sufficient to induce substantial growth delays of both CoLo201 and CoLo205 s.c. tumors in nude mice. This growth delay equates to between 40 and 60% inhibition of tumor protein synthesis as quantified by an in vivo [14C]leucine incorporation assay. Using this technique, it was shown that protein synthesis inhibition persisted for at least 96 h after a single dose of ZD0490. Administration of the same total dose given as daily doses over 5 days did not alter the antitumor efficacy of ZD0490 in either the growth delay or the protein synthesis inhibition assays. The in vitro and in vivo activity of ZD0490 detailed in this paper show that this novel immunotoxin is worthy of clinical evaluation, which is currently under way in the United Kingdom.


Asunto(s)
Neoplasias Colorrectales/terapia , Inmunotoxinas/uso terapéutico , Ricina/uso terapéutico , Animales , Anticuerpos , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Neoplasias Colorrectales/metabolismo , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Neoplasias/biosíntesis , Células Tumorales Cultivadas
7.
Cancer Res ; 53(20): 4881-9, 1993 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-8402676

RESUMEN

Pharmacokinetic studies of etoposide administered at 100-200 mg/m2 to 33 children are described. Twenty-seven studies were performed in children aged < 10 years. Repeat studies were performed in 11 patients. Median pharmacokinetic parameters were as follows: plasma clearance, 26 ml/min/m2; volume of distribution, 4.9 liters/m2; area under the etoposide plasma concentration-time curve (AUC), 3.9 mg/ml x min per 100 mg/m2. Interindividual variability in pharmacokinetic parameters was large (coefficient of variation (CV) = 30, 28, and 27%, respectively) in comparison with intraindividual variability (CV = 12, 14, and 12%, respectively). Variability in AUC was much greater in those patients treated with 150-200 mg/m2 etoposide than with 100 mg/m2 (CV, 35 versus 13%) and was related to variability in renal function and prior exposure to cisplatin. Data from the first 20 studies were used to develop pharmacokinetic monitoring equations which were validated in a further 13 patients. The most accurate equation relies upon the elimination constant of 51Cr-EDTA and a single blood specimen taken at the end of the etoposide infusion. [formula: see text] where K = 51Cr-EDTA elimination rate constant. This equation showed no significant bias, and the predictive error was small with respect to AUC calculated according to a two-compartment model. Predictive error did not increase with increasing AUC, whereas a marked increase in predictive error was seen for dosing according to body surface area. Dosing according to body surface area alone led to marked over- or underexposure to etoposide in 8 patients. Pharmacokinetic monitoring using the equation described would have identified these patients and permitted dose modification. This approach provides an accurate means of monitoring etoposide AUC for administration times of 1-4 h without the need for detailed pharmacokinetic sampling. It will allow a significant reduction in the variability of exposure seen with surface area-based dosing.


Asunto(s)
Etopósido/farmacocinética , Etopósido/uso terapéutico , Neoplasias/tratamiento farmacológico , Adolescente , Neoplasias Encefálicas/tratamiento farmacológico , Niño , Preescolar , Etopósido/sangre , Femenino , Humanos , Lactante , Masculino , Tasa de Depuración Metabólica , Neoplasias/sangre , Neuroblastoma/tratamiento farmacológico , Glándula Pineal , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Sarcoma/tratamiento farmacológico , Neoplasias de los Tejidos Blandos/tratamiento farmacológico , Teratoma/tratamiento farmacológico
8.
Cancer Res ; 44(4): 1693-7, 1984 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-6367971

RESUMEN

cis-Diammine-1,1-cyclobutane dicarboxylate platinum(II) (CBDCA, JM8) is a nonnephrotoxic analogue of cisplatin currently undergoing clinical evaluation. Pharmacokinetic studies have been performed in patients receiving CBDCA (20 to 520 mg/sq m) as a 1-hr infusion without hydration or diuresis. Following the end of the infusion, plasma levels of total platinum and ultrafilterable (Mr less than 50,000) platinum (free platinum) decayed biphasically with first-order kinetics (total platinum t alpha 1/2 = 98 min; t beta 1/2 range, 399 to greater than 1440 min; free platinum t alpha 1/2 = 87 min; t beta 1/2 = 354 min). During the first four hr, binding of platinum to plasma protein was limited (24%), with most of the free platinum in the form of unchanged CBDCA (94%). However, by 24 hr, the majority of platinum was protein bound (87%). The major route of elimination was renal, 65% of the platinum administered being excreted in the urine within 24 hr, with 32% of the dose excreted as unchanged CBDCA. No evidence was found from studies on the renal clearance of free platinum to indicate renal tubular secretion (mean free platinum renal clearance, 69 ml/min). However, the plasma clearance of free platinum did correlate positively with glomerular filtration rates (p = 0.005). None of the pharmacokinetic parameters determined were dose dependent. In vitro studies with plasma and urine demonstrated that, in contrast to cisplatin, CBDCA is a stable complex [t 1/2 - 37 degrees; plasma, 30 hr, and urine (range), 20 to 460 hr]. The differences in the pharmacokinetics of cisplatin and CBDCA may explain why the latter complex is not nephrotoxic.


Asunto(s)
Antineoplásicos/toxicidad , Riñón/efectos de los fármacos , Compuestos Organoplatinos/toxicidad , Carboplatino , Evaluación de Medicamentos , Humanos , Cinética , Neoplasias/tratamiento farmacológico , Compuestos Organoplatinos/sangre , Platino (Metal)/sangre
9.
Cancer Res ; 50(23): 7544-8, 1990 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-2253202

RESUMEN

N10-Propargyl-5,8-dideazafolic acid (CB3717) and 2-desamino-2-methyl-N10-propargyl-5,8-dideazafolic acid (ICI-198,583) are potent folate-based inhibitors of thymidylate synthase. We studied the membrane transport and the growth-inhibitory effects of the two thymidylate synthase inhibitors on human CCRF-CEM leukemia cells with different transport properties for folic acid, reduced folates, and methotrexate (MTX). Membrane transport of [3H]ICI-198,583 can proceed via the high affinity/low capacity reduced folate carrier as supported by findings that (a) uptake of [3H]ICI-198,583 was significantly impaired in CEM cells which have a transport defect for MTX, (b) variants of CEM cells which overproduce the reduced folate carrier system showed a concomitant increase in the uptake of [3H]ICI-198,583 as for [3H]MTX, (c) MTX inhibited transport of [3H]ICI-198,583, and (d) uptake of [3H]ICI-198,583 was inhibited after treatment of CEM cells with an N-hydroxysuccinimide ester of MTX, which is a potent inhibitor of MTX transport. However, a membrane-associated folate-binding protein (FBP) offers another route for entry of CB3717 and ICI-198,583. CEM-FBP cells that have an elevated amount of FBP and do not have a functional reduced folate carrier were 640- and 61-fold more sensitive to CB3717 and ICI-198,583, respectively, compared to control CEM cells expressing the reduced folate/MTX carrier. This high sensitivity was related to a high affinity of the FBP for CB3717 and ICI-198,583 (Kd 2-3 nM), which is only 3-fold lower than for folic acid (Kd 1 nM) but significantly higher than for MTX (Kd 100 nM). Furthermore, after incubation of CEM-FBP cells for 24 h at 10 nM [3H]ICI-198,583, the high affinity binding of the FBP for ICI-198,583 allowed a 600-fold concentrative uptake of [3H]ICI-198,583 and its conversion to polyglutamate forms. These results indicate that multiple folate transport systems may be involved in the uptake of folate-based thymidylate synthase inhibitors.


Asunto(s)
Antineoplásicos/farmacocinética , Ácido Fólico/análogos & derivados , Leucemia/metabolismo , Quinazolinas/farmacocinética , Timidilato Sintasa/antagonistas & inhibidores , Transporte Biológico , División Celular/efectos de los fármacos , Células Cultivadas , Portadores de Fármacos , Ácido Fólico/metabolismo , Ácido Fólico/farmacocinética , Antagonistas del Ácido Fólico/farmacocinética , Humanos , Técnicas In Vitro , Metotrexato
10.
Cancer Res ; 48(22): 6396-403, 1988 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-3263186

RESUMEN

A monoclonal anti-Thy-1.1 antibody (OX7) was coupled to either native or chemically deglycosylated ricin A-chain (dgA) using one of two different cross-linking agents. One cross-linker, N-succinimidyloxycarbonyl-alpha-methyl-alpha-(2-pyridyldithio)tolu ene (SMPT), generates a sterically hindered disulfide bond which is relatively resistant to reduction, whereas the other, 2-iminothiolane hydrochloride, generates an unhindered disulfide bond with greater lability. A two-compartment pharmacokinetic model was used to analyze the blood levels of each immunotoxin and its breakdown product (free antibody) after i.v. injection into mice. Immunotoxins prepared with SMPT broke down in vivo 6.3-fold more slowly than those prepared with 2-iminothiolane hydrochloride, and immunotoxins containing native A-chain were cleared 2- to 3-fold more rapidly from the bloodstream than those containing dgA. As a result, 24 h after injection, 16% of the OX7-SMPT-dgA remained in the blood as compared with 0.4 to 2.5% of the other immunotoxins. Immunotoxins prepared with dgA were about 3-fold more toxic to mice than those prepared with native A-chain, whereas immunotoxins prepared with SMPT were only slightly more toxic than those prepared with 2-iminothiolane hydrochloride. When equivalent toxic doses of the immunotoxins were administered i.v. to mice which had been given injections of Thy-1.1+ AKR-A/2 lymphoma cells, the OX7-SMPT-dgA gave the best antitumor effect. A dose equivalent to one-seventh of the median lethal dose extended the survival time of the animals by the extent expected if 99.999% of the tumor cells had been eradicated. Furthermore, the tumors that did develop in the mice treated with OX7-SMPT-dgA were mutants which were resistant to all the immunotoxins. Some of the mutants were deficient in Thy-1.1 whereas others were not. In conclusion, both the use of the SMPT cross-linker and deglycosylation of the A-chain significantly improve the therapeutic index of the immunotoxins in AKR-A/2 tumor-bearing mice.


Asunto(s)
Antineoplásicos/farmacología , Inmunotoxinas/farmacología , Ricina/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Estabilidad de Medicamentos , Femenino , Glicosilación , Inmunotoxinas/metabolismo , Isoanticuerpos/inmunología , Dosificación Letal Mediana , Linfoma/patología , Linfoma/terapia , Ratones , Ratones Endogámicos AKR , Ratones Endogámicos BALB C , Células Tumorales Cultivadas
11.
Cancer Res ; 48(24 Pt 1): 7072-8, 1988 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-3263899

RESUMEN

Immunotoxins containing the ribosome-inactivating protein, saporin, are very effective antitumor agents but are highly toxic to mice. They induce severe necrotic lesions in the liver parenchyma of the recipients. Such extensive damage to the liver parenchyma is not observed with ricin A-chain immunotoxins even at 5-fold higher dosage. The hepatotoxicity of the saporin immunotoxins was found in the present study to arise from a combination of two effects. First, saporin and saporin immunotoxins were 30- and 6-fold more toxic to primary cultures of mouse liver parenchymal cells than were ricin A-chain and ricin A-chain immunotoxins, respectively. This was despite the fact that the cells bound 4- to 5-fold less saporin or saporin immunotoxins than ricin A-chain or ricin A-chain immunotoxins. The binding of ricin A-chain and its immunotoxin to the cells was mediated through the carbohydrate residues present on the A-chain whereas saporin is not glycosylated and thus must bind to other sites on the cell surface which result in transport of saporin relatively efficiently to the cytosol. The second reason for the hepatotoxic action of the saporin immunotoxin was that it had a longer blood half-life (t 1/2 alpha = 1.1 h; t 1/2 beta = 17.1 h) than the ricin A-chain immunotoxin (t 1/2 = 0.52 h; t 1/2 beta = 9.7 h). Analyses using a two-compartment pharmacokinetic model showed that the two immunotoxins broke down in vivo to give free antibody at a similar rate (t 1/2 = 10-12 h) but that the ricin A-chain immunotoxin was eliminated 11 times more rapidly than the saporin immunotoxin by routes other than breakdown. It was calculated that, in mice given a median lethal dose of saporin immunotoxin, the blood levels of immunotoxin remained above the concentration that killed 50% of parenchymal cells in vitro for more than 48 h. In mice given a median lethal dose of ricin A-chain immunotoxin, the blood levels fell below the concentration that was toxic to parenchymal cells in vitro within 4 h. The longer blood half-life of the saporin immunotoxin may also explain our previous finding that it had antitumor activity superior to that of a ricin A-chain immunotoxin in mice.


Asunto(s)
Inmunotoxinas/farmacocinética , Hígado/efectos de los fármacos , N-Glicosil Hidrolasas , Proteínas de Plantas/toxicidad , Ricina/farmacocinética , Animales , Células Cultivadas , Semivida , Matemática , Ratones , Proteínas de Plantas/farmacocinética , Proteínas Inactivadoras de Ribosomas Tipo 1 , Ricina/toxicidad , Saporinas
12.
Cancer Res ; 52(3): 603-9, 1992 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-1732048

RESUMEN

The pharmacokinetics of the anthrapyrazole CI-941 has been investigated in conjunction with the Phase I evaluation of the drug with the intent of applying a pharmacokinetically guided dose escalation strategy. A starting dose of 5 mg/m2 was chosen, based on one-tenth the 10% lethal dose in mice. Due to the steep dose lethality relationship and nonlinear pharmacokinetics in mice, a target area under the CI-941 plasma concentration x time curve (AUC) of 110 microM x min (i.e., 40% of the mouse 10% lethal dose AUC) was chosen. This AUC was achieved in mice at 40 mg/m2. A total of 37 patients received 74 courses of CI-941 (5 to 55 mg/m2), with 26 patients consenting to pharmacokinetic monitoring. CI-941 was rapidly cleared from plasma, and a triexponential open model could be fitted to the data (t1/2 alpha = 7.6 +/- 2 min, t1/2 beta = 65 +/- 27 min, t1/2 zeta = 21 +/- 9 h). CI-941 was subjected to only limited urinary elimination, accounting for 5.2 +/- 2.8% of the administered dose. Wide interpatient variability in plasma CI-941 clearance at the starting dose and subsequent doses precluded the implementation of a pharmacokinetically guided dose escalation scheme, and the dose was escalated in 5-mg/m2 increments until the maximally tolerated dose was achieved. A number of investigations were performed to study potential reasons for variability in CI-941 clearance. However, CI-941 plasma protein binding (95 +/- 1%) and measures of pretreatment renal (51Cr-EDTA clearance), hepatic (plasma alanine transaminase and alkaline phosphatase levels), or cardiac function (left ventricular ejection fractions) did not relate strongly to CI-941 clearance. In patients treated at 40 mg/m2, the AUC values (156 to 415 microM x min) approximated or exceeded the target AUC. Fifty mg/m2 was the Phase II recommended dose. Further prospective studies are warranted to assess the utility of pharmacokinetically guided dose escalation strategies and to determine whether or not the variability encountered in clearance is unique to CI-941.


Asunto(s)
Antraquinonas/farmacocinética , Antineoplásicos/farmacocinética , Neoplasias/tratamiento farmacológico , Pirazoles/farmacocinética , Pirazolonas , Antraquinonas/uso terapéutico , Antraquinonas/toxicidad , Proteínas Sanguíneas/metabolismo , Radioisótopos de Carbono , Evaluación de Medicamentos , Tasa de Filtración Glomerular/efectos de los fármacos , Humanos , Tasa de Depuración Metabólica , Orosomucoide/metabolismo , Unión Proteica , Pirazoles/uso terapéutico , Pirazoles/toxicidad , Análisis de Regresión , Distribución Tisular
13.
Cancer Res ; 50(23): 7519-26, 1990 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-2253201

RESUMEN

A panel of immunotoxins was constructed by chemically attaching the ribosome-inactivating proteins abrin A chain, ricin A chain, gelonin, and momordin to the monoclonal mouse IgG2a antibody Fib75 by means of a disulfide linkage. All the immunotoxins were toxic in tissue culture to the EJ human bladder carcinoma cell line expressing the antigen recognized by Fib75, inhibiting the incorporation of [3H]leucine by 50% at concentrations between 1 x 10(-10) M and 8 x 10(-10) M. The pharmacokinetics of the immunotoxins in the normal Wistar rat was determined following i.v. administration. The concentrations of intact immunotoxin in serum samples taken at various intervals after injection for up to 24 h were measured by solid-phase enzyme-linked immunosorbent assays specific for each of the four different ribosome-inactivating proteins. The Fib75 immunotoxins were cleared from the circulation with comparable, but not identical, biphasic kinetics best described by a two compartment open pharmacokinetic model. The alpha-phase half-lives of the panel, between 0.35 and 0.71 h, were similar. The beta-phase half-life of Fib75-abrin A chain, 13.3 h, was significantly longer than the beta-phase half-lives of Fib75-ricin A chain, Fib75-gelonin, and Fib75-momordin, between 7.5 and 8.6 h. Fib75-abrin A chain was found to be about 3- to 4-fold more resistant than the other immunotoxins to breakdown by reduction of the disulfide linkage between the A chain and the antibody with glutathione in vitro. This suggests that the longer serum half-life of Fib75-abrin A chain may have been due to greater stability against reduction in vivo. Analysis of serum samples obtained up to 24 h after injection of Fib75-abrin A chain revealed that the chemically intact immunotoxin present in the circulation retained full cytotoxic activity. An abrin A chain immunotoxin made with a different monoclonal mouse IgG2a antibody was also found to be more stable against reduction by glutathione in vitro than an analogous ricin A chain immunotoxin. Thus, abrin A chain may posses unique molecular properties that endow immunotoxins made with this A chain with greater stability in vivo than immunotoxins made with ricin A chain or other ribosome-inactivating proteins.


Asunto(s)
Abrina/uso terapéutico , Inmunotoxinas/farmacocinética , N-Glicosil Hidrolasas , Proteínas de Plantas/uso terapéutico , Ricina/uso terapéutico , Animales , Anticuerpos Monoclonales/uso terapéutico , Antimetabolitos/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunotoxinas/toxicidad , Masculino , Tasa de Depuración Metabólica , Inhibidores de la Síntesis de la Proteína/uso terapéutico , Ratas , Proteínas Inactivadoras de Ribosomas Tipo 1 , Proteínas Inactivadoras de Ribosomas Tipo 2 , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico
14.
J Clin Oncol ; 11(12): 2314-23, 1993 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8246021

RESUMEN

PURPOSE: The aim of this study was to define the pharmacokinetics of carboplatin in children and use the data to develop a pediatric dose formula. It was anticipated that renal function would be a major determinant of carboplatin disposition and the relationship between carboplatin clearance and glomerular filtration rate (GFR) was examined in detail. PATIENTS AND METHODS: Plasma carboplatin pharmacokinetics were measured as ultrafiltrable platinum in 22 patients (5 to 63 kg) following 200 to 1,000 mg/m2 of carboplatin. GFR was measured by the plasma clearance of chromium 51-edathamil (51Cr-EDTA). RESULTS: Carboplatin pharmacokinetics in children were best described in most patients (16 of 22) by a two-compartment model. The dose-normalized area under the plasma carboplatin concentration versus time curve (AUC) ranged from 3.1 to 9.6 mg/mL.min/400 mg/m2 and there was only a weak linear relationship between carboplatin dose and AUC (R2 = .31). There was a significant relationship between absolute carboplatin and 51Cr-EDTA clearances (R2 = .56), but the relationship was weaker (R2 = .28) when both clearances were normalized for body surface area. Carboplatin plasma clearance was predicted by the equation: clearance = GFR (mL/min) + 0.36 x body weight (BW; kg), and a modified form of the adult carboplatin dose formula is proposed: dose (mg) = target AUC x (GFR [mL/min] + [0.36 x BW(kg)]). Two further equations were developed that use the 51Cr-EDTA half-life (t1/2) to calculate the GFR and these may reduce errors resulting from inaccurate measurement of the volume of distribution for 51Cr-EDTA. In patients treated with single-agent carboplatin or carboplatin plus vincristine, there was a significant sigmoidal relationship between AUC and thrombocytopenia (R2 = .56). CONCLUSION: GFR-based carboplatin dosing in children should be feasible and will be evaluated prospectively.


Asunto(s)
Carboplatino/administración & dosificación , Carboplatino/farmacocinética , Neoplasias/metabolismo , Adolescente , Carboplatino/efectos adversos , Niño , Preescolar , Radioisótopos de Cromo , Ácido Edético , Femenino , Tasa de Filtración Glomerular , Humanos , Lactante , Pruebas de Función Renal , Masculino , Neoplasias/tratamiento farmacológico
15.
J Clin Oncol ; 4(8): 1245-52, 1986 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-3734849

RESUMEN

CB3717 is a quinazoline antifolate whose cytotoxic activity is mediated by inhibition of thymidylate synthase (TS). A phase I clinical trial commenced in September 1981 and 99 patients have received 296 treatments. Doses were dissolved in 0.15 mol/L NaHCO3 (pH 9.0) at a concentration of 4 mg/mL infused over one hour or in a total volume of 1 L infused over 12 hours. Doses were repeated every 3 weeks. The starting dose of 140 mg/m2 was escalated to 600 mg/m2. Renal toxicity, detected by a decrease in the 51Cr EDTA clearance, was dose-related and occurred in seven of ten patients receiving greater than 450 mg/m2. Reversible hepatic toxicity often associated with malaise occurred in 223 of 288 assessable courses (77%). Fifty-nine courses (20%) were associated with increases in alanine transaminase (ALT) levels to greater than 2.5 times the upper limit of the normal laboratory range. Increases in alkaline phosphatase levels also occurred, but were less marked. The severity and prevalence of these elevations were unaffected by the duration of the infusion. A self-limiting rash appeared in 12 patients and a radiation recall reaction was seen in two. Leukopenia developed in 17 patients (WBC less than 3 X 10(9)/L), and thrombocytopenia occurred in six patients (platelets less than 100 X 10(9)/L). The mean leucocyte nadir occurred on day 10 and was followed by recovery at 11 to 19 days. Neither the incidence nor the severity of any of these latter toxicities was dose related. The maximum tolerated dose was in the region of 600 mg/m2 with renal toxicity being dose limiting, although the inter-patient variation did not allow a precise definition. Seventy-six patients were evaluable for response. Responses occurred at doses greater than or equal to 200 mg/m2 and were ovary, one complete response (CR), one partial response (PR), seven minor responses (MR) in 30 cases; breast, two PRs and one MR in eight cases; adenocarcinoma of the lung, one MR in 5 cases; mesothelioma, one PR in five cases; and colon, two MRs in four cases. CB3717 has activity in heavily pretreated patients. The recommended phase II dose for good-risk patients is 400 mg/m2 using the one-hour infusion schedule of administration.


Asunto(s)
Antineoplásicos/uso terapéutico , Antagonistas del Ácido Fólico/uso terapéutico , Ácido Fólico/análogos & derivados , Neoplasias/tratamiento farmacológico , Quinazolinas/uso terapéutico , Timidilato Sintasa/antagonistas & inhibidores , Acetilglucosaminidasa/orina , Fosfatasa Ácida/sangre , Alanina Transaminasa/sangre , Antineoplásicos/administración & dosificación , Enfermedad Hepática Inducida por Sustancias y Drogas , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Evaluación de Medicamentos , Antagonistas del Ácido Fólico/administración & dosificación , Tasa de Filtración Glomerular/efectos de los fármacos , Enfermedades Hematológicas/inducido químicamente , Hiperbilirrubinemia/inducido químicamente , Enfermedades Renales/inducido químicamente , Leucil Aminopeptidasa/orina , Neoplasias/sangre , Neoplasias/fisiopatología , Quinazolinas/administración & dosificación , Enfermedades de la Piel/inducido químicamente
16.
J Clin Oncol ; 7(11): 1748-56, 1989 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-2681557

RESUMEN

A dosage formula has been derived from a retrospective analysis of carboplatin pharmacokinetics in 18 patients with pretreatment glomerular filtration rates (GFR) in the range of 33 to 136 mL/min. Carboplatin plasma clearance was linearly related to GFR (r = 0.85, P less than .00001) and rearrangements of the equation describing the correlation gave the dosage formula dose (mg) = target area under the free carboplatin plasma concentration versus time curve (AUC) x (1.2 x GFR + 20). In a prospective clinical and pharmacokinetic study the formula was used to determine the dose required to treat 31 patients (GFR range, 33 to 135 mL/min) with 40 courses of carboplatin. The target AUC was escalated from 3 to 8 mg carboplatin/mL/min. Over this AUC range the formula accurately predicted the observed AUC (observed/predicted ratio 1.24 +/- 0.11, r = 0.886) and using these additional data, the formula was refined. Dose (mg) = target AUC x (GFR + 25) is now the recommended formula. AUC values of 4 to 6 and 6 to 8 mg/mL. min gave rise to manageable hematological toxicity in previously treated and untreated patients, respectively, and hence target AUC values of 5 and 7 mg/mL min are recommended for single-agent carboplatin in these patient groups. Pharmacokinetic modeling demonstrated that the formula was reasonably accurate regardless of whether a one- or two-compartment model most accurately described carboplatin pharmacokinetics, assuming that body size did not influence nonrenal clearance. The validity of this assumption was demonstrated in 13 patients where no correlation between surface area and nonrenal clearance was found (r = .31, P = .30). Therefore, the formula provides a simple and consistent method of determining carboplatin dose in adults. Since the measure of carboplatin exposure in the formula is AUC, and not toxicity, it will not be influenced by previous or concurrent myelosuppressive therapy or supportive measures. The formula is therefore applicable to combination and high-dose studies as well as conventional single-agent therapy, although the target AUC for carboplatin will need to be redefined for combination chemotherapy.


Asunto(s)
Antineoplásicos/administración & dosificación , Riñón/fisiología , Compuestos Organoplatinos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Carboplatino , Relación Dosis-Respuesta a Droga , Tasa de Filtración Glomerular , Humanos , Estudios Prospectivos , Trombocitopenia/inducido químicamente
17.
J Clin Oncol ; 18(21): 3614-21, 2000 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11054434

RESUMEN

PURPOSE: Carboplatin dosing in adults with cancer is based on renal function. The purpose of the current study was to validate a previously developed pediatric carboplatin-dosing formula. PATIENTS AND METHODS: Thirty-eight pediatric patients were randomized to receive a carboplatin dose calculated according to surface area or a renal function-based dosing formula. On the next course of therapy, the alternative dosing method was used for each patient. Carboplatin pharmacokinetics (based on free plasma platinum concentrations) were measured after both courses. RESULTS: The mean observed areas under the carboplatin concentration-versus-time curve (AUCs) after renal function- and surface area-based dosing were 98% and 95% of the target AUCs, respectively. The variation in the observed AUC was significantly less after renal function-based dosing (F test, P =.02), such that 74% of courses had an observed AUC within +/- 20% of the target value, versus 49% for courses after dosing according to surface area. Only one of 22 courses at the center with the most experience with renal function-based dosing was associated with an AUC outside +/- 20% of the target value, versus nine of 22 courses after surface area-based dosing in the same center. There was a relationship (r(2) =.71) between carboplatin AUC and thrombocytopenia in 10 neuroblastoma patients treated with a combination of carboplatin, vincristine, etoposide, and cyclophosphamide. CONCLUSION: Renal function-based carboplatin dosing in children results in more consistent drug exposure than surface area-based drug administration.


Asunto(s)
Antineoplásicos/administración & dosificación , Carboplatino/administración & dosificación , Riñón/fisiología , Adolescente , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Área Bajo la Curva , Superficie Corporal , Carboplatino/efectos adversos , Carboplatino/farmacocinética , Niño , Preescolar , Relación Dosis-Respuesta a Droga , Femenino , Tasa de Filtración Glomerular/fisiología , Humanos , Lactante , Masculino , Estudios Prospectivos , Reproducibilidad de los Resultados
18.
J Clin Oncol ; 16(3): 1131-41, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9508200

RESUMEN

PURPOSE: A phase I, multicenter trial of the thymidylate synthase (TS) inhibitor THYMITAQ (nolatrexed dihydrochloride; Agouron Pharmaceuticals, Inc, San Diego, CA) given by 5-day continuous infusion was performed to establish the maximum-tolerated dose (MTD) and to investigate pharmacokinetics, pharmacodynamics, and antitumor effects. METHODS: In vitro and in vivo preclinical studies demonstrated increased activity with prolonged nolatrexed exposure. In 32 patients, nolatrexed was given as a 5-day infusion at 96 to 1,040 mg/m2/d for 5 days. Pharmacokinetics were determined from high-performance liquid chromatography (HPLC) analyses of plasma and urine. In addition to studying toxicity, plasma deoxyuridine (UdR) elevations were measured as a marker of TS inhibition. RESULTS: The MTD was 904 mg/m2/d for 5 days and the recommended phase II dose is 800 mg/m2/d for 5 days. The dose-limiting toxicity was neutropenia with clinically significant thrombocytopenia and mucositis. These antiproliferative toxicities of nolatrexed were predictable and reversible. A partial response that lasted 3 months occurred in a patient with metastatic colorectal cancer. Pharmacokinetics were nonlinear, with the median plasma clearance (CI) decreasing from 151 mL/min/m2 (range, 124 to 211) at 96 mg/m2/d for 5 days to 49 mL/min/m2 (range, 30 to 84) at 768 mg/ m2/d for 5 days. The half-life (t1/2) was 173 minutes (range, 43 to 784) and 18% (range, 9% to 35%) of the dose was excreted unchanged in the urine. Plasma UdR increased, but returned to pretreatment levels after the end of infusion. Hematologic toxicity was significantly related to nolatrexed plasma concentrations and dose. CONCLUSION: Nolatrexed can be safely administered to patients at a dose of 800 mg/m2/d over 5 days by continuous intravenous infusion and this schedule is associated with antitumor effects. The phase II evaluation of nolatrexed is ongoing.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Antagonistas del Ácido Fólico/farmacología , Neoplasias/tratamiento farmacológico , Quinazolinas/farmacología , Timidilato Sintasa/antagonistas & inhibidores , Adulto , Anciano , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/farmacocinética , Estudios de Evaluación como Asunto , Femenino , Antagonistas del Ácido Fólico/administración & dosificación , Antagonistas del Ácido Fólico/farmacocinética , Humanos , Masculino , Ratones , Persona de Mediana Edad , Quinazolinas/administración & dosificación , Quinazolinas/farmacocinética , Células Tumorales Cultivadas
19.
Clin Cancer Res ; 1(12): 1479-86, 1995 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9815947

RESUMEN

(6R)-5,10-Dideaza-5,6,7,8-tetrahydrofolic acid (lometrexol) is an antipurine antifolate which selectively inhibits glycinamide ribonucleotide formyltransferase. Lometrexol pharmacokinetics were evaluated in 17 patients (32 courses) as part of a Phase I study in which folic acid supplementation was used to improve tolerance to the drug, its clinical utility being previously limited by severe cumulative toxicity. Lometrexol was administered as an i.v. bolus every 4 weeks at a starting dose of 12 mg/m2, with subsequent interpatient dose escalation to 16, 30, and 45 mg/m2. p.o. folic acid (5 mg/day) was given for 7 days before and 7 days after lometrexol administration. The disposition of total lometrexol in plasma was best described by a biexponential model for data acquired up to 12 h after drug administration, although triexponential plasma pharmacokinetics were often found to give a more adequate description when data were available at later time intervals (24 h and greater). Mean plasma half-lives (+ SD) for model-dependent analysis were t1/2alpha 19 +/- 7 min, t1/2beta 256 +/- 96 min, and t1/2gamma (where measurable) 1170 +/- 435 min. Lometrexol area under plasma concentration versus time curve was proportional to the dose administered. Moderate plasma protein binding of lometrexol was evident (78 +/- 3%) with an inverse linear relationship between fraction of unbound lometrexol and the concentration of serum albumin. The volume of distribution of lometrexol at steady state was between 4.7 and 15.8 l/m2. Renal elimination of lometrexol, studied in 19 patients (21 courses), was considerable, accounting for 56 +/- 17% of the total dose administered within 6 h of treatment, and 85 +/- 16% within 24 h of treatment. These recoveries of unchanged lometrexol indicate that the drug does not appear to undergo appreciable systemic metabolism at the range of concentrations studied. Lometrexol pharmacokinetics were also examined in seven patients who received 45 or 60 mg/m2 lometrexol as part of a separate study of the drug given with folinic acid rescue 5-7 days after treatment. No marked differences were evident in lometrexol plasma half-lives, plasma clearance, or the extent of plasma protein binding, indicating that there is not a pronounced pharmacokinetic interaction between lometrexol and folic acid.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacocinética , Ácido Fólico/administración & dosificación , Tetrahidrofolatos/farmacocinética , Adulto , Anciano , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/sangre , Antimetabolitos Antineoplásicos/orina , Área Bajo la Curva , Proteínas Sanguíneas/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/sangre , Neoplasias/orina , Tetrahidrofolatos/administración & dosificación , Tetrahidrofolatos/sangre , Tetrahidrofolatos/orina
20.
Clin Cancer Res ; 6(1): 271-7, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10656458

RESUMEN

The antifolate LY309887 is a specific glycinamide ribonucleotide formyltransferase inhibitor that blocks de novo purine synthesis and produces a depletion of purine nucleotides. The activity of LY309887 in six human tumor cell lines has been examined by growth inhibition and clonogenic assay after continuous exposure for three cell doubling times and by ATP depletion at 24 h. Three cell lines (CCRF-CEM, MCF7, and GC3) were sensitive to LY309887-induced growth inhibition (IC50: 5.6-8.1 nM), whereas the other cell lines (COR-L23, T-47D, and A549) were comparatively resistant (IC50: 36-55 nM). Sensitivity to LY309887 cytotoxicity was consistent with sensitivity to growth inhibition in four of five cell lines tested (MCF7/GC3: 0.01% survival and COR-L23/T-47D: 1-5% survival at 100 nM LY309887). LY309887-induced ATP depletion was measured by luciferase-based ATP assay and confirmed by high performance liquid chromatography measurements. There was a linear relationship between ATP depletion and growth inhibition when data were analyzed for all six cell lines (r2 = 0.93; P < 0.0001). Depletion of 24-h cellular ATP concentrations to < 1 mM was associated with both cell growth inhibition and cytotoxicity in all cell lines studied. In conclusion, cellular ATP depletion induced by LY309887 can be used to predict growth inhibition and cytotoxicity in human tumor cells.


Asunto(s)
Adenosina Trifosfato/metabolismo , Antineoplásicos/toxicidad , División Celular/efectos de los fármacos , Inhibidores Enzimáticos/toxicidad , Tetrahidrofolatos/toxicidad , Neoplasias de la Mama , Tamaño de la Célula/efectos de los fármacos , Femenino , Humanos , Transferasas de Hidroximetilo y Formilo/antagonistas & inhibidores , Neoplasias Pulmonares , Fosforribosilglicinamida-Formiltransferasa , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA