Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
ACS Infect Dis ; 9(11): 2141-2159, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37828912

RESUMEN

The lipoteichoic acid (LTA) biosynthesis pathway has emerged as a promising antimicrobial therapeutic target. Previous studies identified the 1,3,4 oxadiazole compound 1771 as an LTA inhibitor with activity against Gram-positive pathogens. We have succeeded in making six 1771 derivatives and, through subsequent hit validation, identified the incorporation of a pentafluorosulfanyl substituent as central in enhancing activity. Our newly described derivative, compound 13, showed a 16- to 32-fold increase in activity compared to 1771 when tested against a cohort of multidrug-resistant Staphylococcus aureus strains while simultaneously exhibiting an improved toxicity profile against mammalian cells. Molecular techniques were employed in which the assumed target, lipoteichoic acid synthase (LtaS), was both deleted and overexpressed. Neither deletion nor overexpression of LtaS altered 1771 or compound 13 susceptibility; however, overexpression of LtaS increased the MIC of Congo red, a previously identified LtaS inhibitor. These data were further supported by comparing the docking poses of 1771 and derivatives in the LtaS active site, which indicated the possibility of an additional target(s). Finally, we show that both 1771 and compound 13 have activity that is independent of LtaS, extending to cover Gram-negative species if the outer membrane is first permeabilized, challenging the classification that these compounds are strict LtaS inhibitors.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Animales , Antibacterianos/química , Mamíferos , Oxadiazoles/farmacología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus
2.
J Liposome Res ; 21(1): 70-80, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20528623

RESUMEN

Liposomes, such as pegylated-liposomal CKD-602 (S-CKD602), undergo catabolism by macrophages and dendritic cells (DCs) of the reticuloendothelial system (RES). The relationship between plasma and tumor disposition of S-CKD602 and RES was evaluated in mice bearing A375 melanoma or SKOV-3 ovarian xenografts. Area under the concentration-time curves (AUCs) of liposomal encapsulated, released, and sum total (encapsulated + released) CKD-602 in plasma, tumor, and tumor extracellular fluid (ECF) were estimated. A375 and SKOV-3 tumors were stained with cd11b and cd11c antibodies as measures of macrophages and DC. The plasma disposition of S-CKD602 was similar in both xenograft models. The ratio of tumor sum total AUC to plasma sum total AUC was 1.7-fold higher in mice bearing human SKOV-3 xenografts, compared with A375. The ratio of tumor ECF AUC to tumor sum total AUC was 2-fold higher in mice bearing human SKOV-3 xenografts, compared with A375. The staining of cd11c was 4.5-fold higher in SKOV-3, compared with A375 (P < 0.0001). The increased tumor delivery and release of CKD-602 from S-CKD602 in the ovarian xenografts, compared with the melanoma xenografts, was consistent with increased cd11c staining, suggesting that variability in the RES may affect the tumor disposition of liposomal agents.


Asunto(s)
Camptotecina/análogos & derivados , Sistema Mononuclear Fagocítico/efectos de los fármacos , Inhibidores de Topoisomerasa I/farmacocinética , Animales , Área Bajo la Curva , Camptotecina/farmacocinética , Camptotecina/farmacología , Cromatografía Liquida , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Espectrometría de Masas , Ratones , Inhibidores de Topoisomerasa I/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Clin Cancer Res ; 14(11): 3529-35, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18519786

RESUMEN

PURPOSE: In vivo, 2',2'-difluoro-2'-deoxycytidine (dFdC) is rapidly inactivated by gut and liver cytidine deaminase (CD) to 2',2'-difluoro-2'-deoxyuridine (dFdU). Consequently, dFdC has poor oral bioavailability and is administered i.v., with associated costs and limitations in administration schedules. 3,4,5,6-Tetrahydrouridine (THU) is a potent CD inhibitor with a 20% oral bioavailability. We investigated the ability of THU to decrease elimination and first-pass effect by CD, thereby enabling oral dosing of dFdC. EXPERIMENTAL DESIGN: A liquid chromatography-tandem mass spectrometry assay was developed for plasma dFdC and dFdU. Mice were dosed with 100 mg/kg dFdC i.v. or orally with or without 100 mg/kg THU i.v. or orally. At specified times between 5 and 1,440 min, mice (n = 3) were euthanized. dFdC, dFdU, and THU concentrations were quantitated in plasma and urine. RESULTS: THU i.v. and orally produced concentrations >4 microg/mL for 3 and 2 h, respectively, whereas concentrations of >1 microg/mL have been associated with near-complete inhibition of CD in vitro. THU i.v. decreased plasma dFdU concentrations but had no effect on dFdC plasma area under the plasma concentration versus time curve after i.v. dFdC dosing. Both THU i.v. and orally substantially increased oral bioavailability of dFdC. Absorption of dFdC orally was 59%, but only 10% passed liver and gut CD and eventually reached the systemic circulation. Coadministration of THU orally increased dFdC oral bioavailability from 10% to 40%. CONCLUSIONS: Coadministration of THU enables oral dosing of dFdC and warrants clinical testing. Oral dFdC treatment would be easier and cheaper, potentially prolong dFdC exposure, and enable exploration of administration schedules considered impractical by the i.v. route.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacocinética , Desoxicitidina/análogos & derivados , Tetrahidrouridina/farmacocinética , Administración Oral , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Área Bajo la Curva , Disponibilidad Biológica , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacocinética , Interacciones Farmacológicas , Masculino , Ratones , Tetrahidrouridina/administración & dosificación , Gemcitabina
4.
Cancer Chemother Pharmacol ; 62(3): 417-26, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17957368

RESUMEN

PURPOSE: We evaluated the antitumor activity of two different schedules of docetaxel and 9-nitrocamptothecin (9NC) in mice bearing human SKOV-3 ovarian carcinoma xenografts and evaluated the plasma, tissue, and tumor disposition of each agent alone and in combination. EXPERIMENTAL DESIGN: The following treatment groups were evaluated: (1) docetaxel 10 mg/kg IV on days 0 and 7; (2) 9NC 0.67 mg/kg PO qdx5dx2wk; (3) 9NC 0.67 mg/kg PO qdx5dx2wk in combination with docetaxel 10 mg/kg IV on days 0 and 7; and (4) 9NC 0.67 mg/kg PO qdx5dx2wk in combination with docetaxel 10 mg/kg IV on days 4 and 11; (5) vehicle controls for each agent; and (6) no treatment controls. RESULTS: All treatment regimens produced significant antitumor activity as compared with control groups (P < 0.05). Docetaxel administered on days 0 and 7 or on days 4 and 11 in combination with 9NC resulted in similar antitumor activity (P > 0.05). High docetaxel concentrations in tumor were maintained at late time points as compared with plasma and tissues with the retention of docetaxel at 24 h being 132-fold and 15-fold higher in tumor than in plasma and liver, respectively. After administration of 9NC alone, the ratio of the 9-aminocamptothecin (9AC) area under the concentration versus time curve (AUC) to 9NC AUC in plasma and tumor was 0.15 and 1.34, respectively. CONCLUSIONS: The combination of docetaxel and 9NC was effective against SKOV-3 xenografts. The lack of a difference in sequence-dependent antitumor activity may reflect the sensitivity of the SKOV-3 xenograft to 9NC. The factors associated with tumor-specific retention of docetaxel and the ratio of 9NC to 9AC in tumors is unknown.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias Ováricas/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/sangre , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Área Bajo la Curva , Camptotecina/análogos & derivados , Camptotecina/sangre , Camptotecina/farmacocinética , Camptotecina/uso terapéutico , Docetaxel , Femenino , Humanos , Ratones , Ratones Endogámicos , Trasplante de Neoplasias , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Taxoides/sangre , Taxoides/farmacocinética , Taxoides/uso terapéutico , Factores de Tiempo , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Chemother Pharmacol ; 62(3): 457-64, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18008070

RESUMEN

Cytidine analogues such as cytosine arabinoside, gemcitabine, decitabine, 5-azacytidine, 5-fluoro-2'-deoxycytidine and 5-chloro-2'-deoxycytidine undergo rapid catabolism by cytidine deaminase (CD). 3,4,5,6-tetrahydrouridine (THU) is a potent CD inhibitor that has been applied preclinically and clinically as a modulator of cytidine analogue metabolism. However, THU pharmacokinetics has not been fully characterized, which has impaired the optimal preclinical evaluation and clinical use of THU. Therefore, we characterized the THU pharmacokinetics and bioavailability in mice. Mice were dosed with THU iv (100 mg/kg) or po (30, 100, or 300 mg/kg). Plasma and urine THU concentrations were quantitated with a validated LC-MS/MS assay. Plasma pharmacokinetic parameters were calculated compartmentally and non-compartmentally. THU, at 100 mg/kg iv had a 73 min terminal half-life and produced plasma THU concentrations >1 microg/ml, the concentration shown to effectively block deamination, for 4 h. Clearance was 9.1 ml/min/kg, and the distribution volume was 0.95 l/kg. Renal excretion accounted for 36-55% of the THU dose. A three-compartment model fit the iv THU data best. THU, at 100 mg/kg po, produced a concentration versus time profile with a plateau of approximately 10 mug/ml from 0.5-3 h, followed by a decline with an 85 min half-life. The oral bioavailability of THU was approximately 20%. The 20% oral bioavailability of THU is sufficient to produce and sustain, for several hours, plasma concentrations that inhibit CD. This suggests the feasibility of using THU to decrease elimination and first-pass metabolism of cytidine analogues by CD. THU pharmacokinetics are now being evaluated in humans.


Asunto(s)
Citidina Desaminasa/antagonistas & inhibidores , Inhibidores Enzimáticos , Tetrahidrouridina , Administración Oral , Animales , Disponibilidad Biológica , Cromatografía Liquida , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Semivida , Inyecciones Intravenosas , Masculino , Ratones , Ratones Endogámicos , Unión Proteica , Espectrometría de Masas en Tándem , Tetrahidrouridina/sangre , Tetrahidrouridina/farmacocinética , Tetrahidrouridina/farmacología
6.
Clin Cancer Res ; 13(23): 7217-23, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18056203

RESUMEN

PURPOSE: S-CKD602 is a STEALTH liposomal formulation of CKD-602, a camptothecin analogue. The cytotoxicity of camptothecin analogues is related to the duration of exposure in the tumor. STEALTH liposomal formulations contain lipid conjugated to methoxypolyethylene glycol and have been designed to prolong drug circulation time, increase tumor delivery, and improve the therapeutic index. For STEALTH liposomal formulations of anticancer agents to achieve antitumor effects, the active drug must be released into the tumor extracellular fluid (ECF). EXPERIMENTAL DESIGN: S-CKD602 at 1 mg/kg or nonliposomal CKD-602 at 30 mg/kg was administered once via tail vein to mice bearing A375 human melanoma xenografts. Mice (n = 3 per time point) were euthanized at 0.083 to 24 h, 48 h, and 72 h after S-CKD02 and from 0.083 to 24 h after nonliposomal CKD-602. Plasma samples were processed to measure encapsulated, released, and sum total (encapsulated plus released) CKD-602, and tumor and tissue samples were processed to measure sum total CKD-602. Microdialysis samples of tumor ECF were obtained from 0 to 2 h, 4 to 7 h, and 20 to 24 h after nonliposomal CKD-602 and from 0 to 2 h, 24 to 27 h, 48 to 51 h, and 72 to 75 h after S-CKD602. A liquid chromatography-mass spectrometry assay was used to measure the total (sum of lactone and hydroxyl acid) CKD-602. The area under the concentration-versus-time curves (AUC) from 0 to infinity and time >1 ng/mL in tumor were estimated. RESULTS: For S-CKD602, the CKD-602 sum total AUC in plasma and tumor and the CKD-602 AUC in tumor ECF were 201,929, 13,194, and 187 ng/mL h, respectively. For S-CKD602, 82% of CKD-602 remains encapsulated in plasma. For nonliposomal CKD-602, the CKD-602 AUC in plasma and tumor and the CKD-602 AUC in tumor ECF were 9,117, 11,661, and 639 ng/mL.h, respectively. The duration of time the CKD-602 concentration was >1 ng/mL in tumor ECF after S-CKD602 and nonliposomal CKD-602 was >72 and approximately 20 h, respectively. For S-CKD602, the CKD-602 sum total exposure was 1.3-fold higher in fat as compared with muscle. The ratio of CKD-602 sum total exposure in fat to muscle was 3.8-fold higher after administration of S-CKD602 compared with nonliposomal CKD-602. CONCLUSION: S-CKD602 provides pharmacokinetic advantages in plasma, tumor, and tumor ECF compared with nonliposomal CKD-602 at 1/30th of the dose, which is consistent with the improved antitumor efficacy of S-CKD602 in preclinical studies. The distribution of S-CKD602 is greater in fat compared with muscle whereas the distribution of nonliposomal CKD-602 is greater in muscle compared with fat. These results suggest that the body composition of a patient may affect the disposition of S-CKD602 and released CKD-602.


Asunto(s)
Camptotecina/análogos & derivados , Liposomas/farmacocinética , Melanoma/metabolismo , Polietilenglicoles/farmacocinética , Animales , Camptotecina/administración & dosificación , Camptotecina/sangre , Camptotecina/farmacocinética , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Líquido Extracelular/metabolismo , Femenino , Humanos , Liposomas/administración & dosificación , Melanoma/sangre , Melanoma/tratamiento farmacológico , Ratones , Ratones SCID , Microdiálisis/métodos , Polietilenglicoles/administración & dosificación , Distribución Tisular , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Anticancer Res ; 27(5A): 3067-73, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17970046

RESUMEN

Cdc25 protein phosphatases are regulators of cyclin-dependent kinases and are often highly expressed in human malignancies. Few small molecule inhibitors of the Cdc25 phosphatase family have been identified and little is known about their disposition, metabolism or efficacy in xenograft models. In this study, the efficacy, pharmacokinetics, and metabolism of a potent quinolinedione Cdc25 phosphatase inhibitor, DA3003-1, in mice was examined. DA3003-1 inhibited the growth of subcutaneous human colon HT29 xenografts in SCID mice. After a single i.v. dose of 5 mg/kg, DA3003-1 was not detectable in plasma or tissues beyond 5 min. In vitro studies showed that DA3003-1 was rapidly dechlorinated and conjugated to glutathione. Following DA3003-1 treatment of tumor-bearing SCID mice, reduced glutathione concentrations in HT29 tumor were decreased to a greater extent and remained decreased for longer than the reduced glutathione concentrations in liver and kidneys. These studies suggest that the minimal antitumor activity of DA3003-1 in mice may be due to its rapid metabolism.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Quinolonas/farmacología , Quinonas/farmacología , Fosfatasas cdc25/antagonistas & inhibidores , Animales , Antineoplásicos/sangre , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Línea Celular Tumoral , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/toxicidad , Femenino , Glutatión/metabolismo , Glutatión Transferasa/metabolismo , Células HT29 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Quinolonas/sangre , Quinolonas/farmacocinética , Quinolonas/toxicidad , Quinonas/sangre , Quinonas/farmacocinética , Quinonas/toxicidad , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Clin Cancer Res ; 12(24): 7483-91, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17138702

RESUMEN

PURPOSE: In vivo, 5-fluoro-2'-deoxycytidine (FdCyd) is rapidly and sequentially converted to 5-fluoro-2'-deoxyuridine, 5-fluorouracil, and 5-fluorouridine. The i.v. combination of FdCyd and 3,4,5,6-tetrahydrouridine (THU), a cytidine deaminase (CD) inhibitor that blocks the first metabolic step in FdCyd catabolism, is being investigated clinically for its ability to inhibit DNA methyltransferase. However, the full effects of THU on FdCyd metabolism and pharmacokinetics are unknown. We aimed to characterize the pharmacokinetics, metabolism, and bioavailability of FdCyd with and without THU in mice. EXPERIMENTAL DESIGN: We developed a sensitive high-performance liquid chromatography tandem mass spectrometry assay to quantitate FdCyd and metabolites in mouse plasma. Mice were dosed i.v. or p.o. with 25 mg/kg FdCyd with or without coadministration of 100 mg/kg THU p.o. or i.v. RESULTS: The oral bioavailability of FdCyd alone was approximately 4%. Coadministration with THU increased exposure to FdCyd and decreased exposure to its metabolites; i.v. and p.o. coadministration of THU increased exposure to p.o. FdCyd by 87- and 58-fold, respectively. FdCyd exposure after p.o. FdCyd with p.o. THU was as much as 54% that of i.v. FdCyd with i.v. THU. CONCLUSIONS: FdCyd is well absorbed but undergoes substantial first-pass catabolism by CD to potentially toxic metabolites that do not inhibit DNA methyltransferase. THU is sufficiently bioavailable to reduce the first-pass effect of CD on FdCyd. Oral coadministration of THU and FdCyd is a promising approach that warrants clinical testing because it may allow maintaining effective FdCyd concentrations on a chronic basis, which would be an advantage over other DNA methyltransferase inhibitors that are currently approved or in development.


Asunto(s)
Metilasas de Modificación del ADN/antagonistas & inhibidores , Desoxicitidina/análogos & derivados , Animales , Antimetabolitos Antineoplásicos/sangre , Antimetabolitos Antineoplásicos/metabolismo , Antimetabolitos Antineoplásicos/farmacocinética , Antimetabolitos Antineoplásicos/orina , Disponibilidad Biológica , Desoxicitidina/sangre , Desoxicitidina/metabolismo , Desoxicitidina/farmacocinética , Desoxicitidina/orina , Relación Dosis-Respuesta a Droga , Masculino , Tasa de Depuración Metabólica , Ratones , Modelos Biológicos
9.
Clin Cancer Res ; 12(19): 5826-33, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17020990

RESUMEN

PURPOSE: To elucidate the in vivo metabolic fate of zebularine (NSC 309132), a DNA methyltransferase inhibitor proposed for clinical evaluation in the treatment of cancer. EXPERIMENTAL DESIGN: Male, CD(2)F(1) mice were dosed i.v. with 100 mg/kg 2-[(14)C]zebularine. At specified times between 5 and 1,440 minutes, mice were euthanized. Plasma, organs, carcass, urine, and feces were collected and assayed for total radioactivity. Plasma and urine were also analyzed for zebularine and its metabolites with a previously validated high-pressure liquid chromatography assay. A similar experiment was done with 2-[(14)C]uridine, the proposed primary metabolite of zebularine. RESULTS: Maximum plasma concentrations were 462, 306, 33.6, 21.7, and 11.5 mumol/L for total radioactivity, zebularine, uridine, uracil (each at 5 minutes), and dihydrouracil (at 15 minutes), respectively. Total radioactivity, zebularine, uridine, uracil, and dihydrouracil were rapidly eliminated from plasma, and after 45 minutes, none of the individual compounds could be quantitated by high-pressure liquid chromatography. Plasma data were consistent with sequential conversion of zebularine to uridine, uracil, and dihydrouracil. 2-Pyrimidinone was not observed. Prolonged retention of radioactivity, at concentrations higher than in plasma, was observed in tissues. Recovery of given radioactivity in urine (30.3% of dose), feces (0.4% of dose), cage wash (7.9% of dose), and tissues and carcass (6.1% of dose) after 24 hours implied that up to 55% of radioactivity was expired as (14)CO(2). Comparison of zebularine and uridine pharmacokinetic data indicated that approximately 40% of the zebularine dose was converted to uridine. CONCLUSIONS: Zebularine is extensively and rapidly metabolized into endogenous compounds that are unlikely to have effects at the concentrations observed.


Asunto(s)
Citidina/análogos & derivados , Metilasas de Modificación del ADN/antagonistas & inhibidores , Animales , Cromatografía Líquida de Alta Presión , Citidina/farmacocinética , Masculino , Ratones , Uracilo/análogos & derivados , Uracilo/sangre , Uridina/sangre
10.
J Photochem Photobiol B ; 88(2-3): 90-104, 2007 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-17604637

RESUMEN

Efficient design of anti-cancer treatments involving radiation- and photo-sensitizing therapeutics requires knowledge of tissue-specific drug concentrations. This study investigates the use of the optical pharmacokinetic system (OPS) to measure concentrations of the anti-cancer agent motexafin gadolinium (MGd) in mouse tissues noninvasively and nondestructively using elastic-scattering spectroscopy. The magnitude of MGd absorbance was quantitated by integration of the MGd peak absorbance area, and MGd concentrations were estimated by comparison with standard curves that were validated by high performance liquid chromatography (HPLC). In tissue-simulating phantoms in vitro, MGd peak absorbance area correlated with MGd concentration. Female C.B-17 SCID mice, bearing subcutaneous MDA-MB-231 human breast cancer xenografts, were dosed with 23 mg/kg MGd i.v. At specific times between 5 min and 24h after dosing, noninvasive OPS measurements were made on skin overlaying the subcutaneous tumor and skin on the opposite flank in vivo, and following exsanguination, nondestructive measurements were made on tumor, skin, and internal tissues in situ. OPS measurements on tissues in vivo detected MGd present in both tissue and blood perfusing the tissue. Both the OPS and the HPLC detected selective localization of MGd in malignant tissues compared with surrounding non-malignant tissues, and neither technique detected MGd in brain tissue. Comparison of MGd concentrations measured by HPLC and OPS is complicated by mismatch between measured tissue volumes, heterogeneous spatial distribution of MGd in tissues, and blood-localized MGd at early time points. Tumor-specific MGd concentrations measured by HPLC correlated with those measured by OPS in vivo and in situ. Best fit lines to the concentration estimates (forced through zero) had slopes of 0.900 and 1.185, respectively; however, the variability was significant (r(2)=0.477 and 0.269). The clinical utility of the OPS to quantitate MGd concentrations remains to be validated.


Asunto(s)
Antineoplásicos/uso terapéutico , Cromatografía Líquida de Alta Presión/métodos , Metaloporfirinas/uso terapéutico , Fármacos Fotosensibilizantes/uso terapéutico , Análisis Espectral/métodos , Animales , Antineoplásicos/farmacocinética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Neoplasias de la Mama/cirugía , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Metaloporfirinas/farmacocinética , Ratones , Fármacos Fotosensibilizantes/farmacocinética , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/cirugía , Factores de Tiempo , Distribución Tisular , Trasplante Heterólogo/métodos
11.
J Chromatogr B Analyt Technol Biomed Life Sci ; 831(1-2): 147-55, 2006 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-16364698

RESUMEN

The metabolism of zebularine (NSC 309132), a novel agent that inhibits DNA methyltransferases, is still uncharacterized. To examine the in vivo metabolism of zebularine, an analytical method was developed and validated (based on FDA guidelines) to quantitate 2-[(14)C]-zebularine and its major metabolites in murine plasma. Zebularine and its metabolites uridine, uracil and dihydrouracil were baseline-separated based on hydrophilic interaction chromatography by using an amino column. The assay was accurate and precise in the concentration ranges of 5.0-100 microg/mL for zebularine, 2.5-50 microg/mL for uridine, 1.0-10 microg/mL for uracil and 0.5-5.0 microg/mL for dihydrouracil. This assay is being used to quantitate zebularine and its metabolites in ongoing pharmacokinetic studies of zebularine.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Citidina/análogos & derivados , Metilasas de Modificación del ADN/antagonistas & inhibidores , Animales , Radioisótopos de Carbono , Citidina/sangre , Citidina/metabolismo , Estabilidad de Medicamentos , Masculino , Ratones , Ratones Endogámicos , Sistemas en Línea , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Organismos Libres de Patógenos Específicos , Espectrofotometría Ultravioleta , Uracilo/análogos & derivados , Uracilo/sangre , Uridina/sangre
12.
Clin Cancer Res ; 11(13): 4867-74, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16000585

RESUMEN

9-Nitrocamptothecin has completed phase III studies in patients with newly diagnosed and refractory pancreatic cancer; however, the optimal 9-nitrocamptothecin treatment regimen is unclear. We used an intermittent schedule of 9-nitrocamptothecin to evaluate the relationship between plasma exposure of 9-nitrocamptothecin and its 9-aminocamptothecin metabolite and antitumor response in mice bearing human colon carcinoma xenografts. 9-Nitrocamptothecin was given orally at 0.44, 0.67, or 1.0 mg/kg/d qd x 5d x 2 weeks repeated q 4 weeks for two cycles to female C.B-17 SCID mice bearing HT29 or ELC2 human colon xenografts. Pharmacokinetic studies were done after oral administration of 0.67 mg/kg x 1. Serial samples were obtained and 9-nitrocamptothecin and 9-aminocamptothecin lactone concentrations in plasma were determined by high-performance liquid chromatography analysis with fluorescence detection. The areas under plasma concentration versus time curve (AUC) from 0 to infinity for 9-nitrocamptothecin and 9-aminocamptothecin were calculated. The antitumor activity of 9-nitrocamptothecin was dose-dependent in both colon xenografts. At all doses, 9-nitrocamptothecin treatment resulted in significant antitumor activity in both xenografts compared with vehicle-treated and control groups and achieved levels of tumor regression that met criteria (minimum %T/C < or = 40%) for antitumor activity. In mice bearing HT29 xenografts, the 9-nitrocamptothecin and 9-aminocamptothecin lactone AUCs after administration of 9-nitrocamptothecin at 0.67 mg/kg were 41.3 and 5.7 ng/mL h, respectively. The responses seen in these xenograft models occurred at systemic exposures that are tolerable in adult patients. These results suggest that the intermittent schedule of 9-nitrocamptothecin may be an active regimen in patients with colorectal carcinoma.


Asunto(s)
Antineoplásicos/farmacocinética , Camptotecina/análogos & derivados , Neoplasias del Colon/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Antineoplásicos/sangre , Antineoplásicos/uso terapéutico , Área Bajo la Curva , Camptotecina/sangre , Camptotecina/farmacocinética , Camptotecina/uso terapéutico , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Femenino , Células HT29 , Humanos , Ratones , Ratones SCID , Resultado del Tratamiento
13.
Clin Cancer Res ; 11(10): 3862-8, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15897587

RESUMEN

PURPOSE: Zebularine is a DNA methyltransferase inhibitor proposed for clinical evaluation. EXPERIMENTAL DESIGN: We developed a liquid chromatography/mass spectrometry assay and did i.v. and oral studies in mice, rats, and rhesus monkeys. RESULTS: In mice, plasma zebularine concentrations declined with terminal half-lives (t(1/2)) of 40 and 91 minutes after 100 mg/kg i.v. and 1,000 mg/kg given orally, respectively. Zebularine plasma concentration versus time curves (area under the curve) after 100 mg/kg i.v. and 1,000 mg/kg given orally were 7,323 and 4,935 mug/mL min, respectively, corresponding to a total body clearance (CL(tb)) of 13.65 mL/min/kg, apparent total body clearance (CL(app)) of 203 mL/min/kg, and oral bioavailability of 6.7%. In rats, plasma zebularine concentrations declined with t(1/2) of 363, 110, and 126 minutes after 50 mg/kg i.v., 250 mg/kg given orally, and 500 mg/kg given orally, respectively. Zebularine areas under the curve after 50 mg/kg i.v., 250 mg/kg given orally, and 500 mg/kg given orally were 12,526, 1,969, and 7,612 mug/mL min, respectively, corresponding to a CL(tb) of 3.99 mL/min/kg for 50 mg/kg i.v. and CL(app) of 127 and 66 mL/min/kg for 250 and 500 mg/kg given orally, respectively. Bioavailabilities of 3.1% and 6.1% were calculated for the 250 and 500 mg/kg oral doses, respectively. In monkeys, zebularine t(1/2) was 70 and 150 minutes, CL(tb) was 3.55 and 10.85 mL/min/kg after i.v. administration, and CL(app) was 886 and 39,572 mL/min/kg after oral administration of 500 and 1,000 mg/kg, respectively. Zebularine oral bioavailability was <1% in monkeys. Interspecies scaling produced the following relationship: CL(tb) = 6.46(weight(0.9)). CONCLUSIONS: Zebularine has limited oral bioavailability. Interspecies scaling projects a CL(tb) of 296 mL/min in humans.


Asunto(s)
Citidina/análogos & derivados , Citidina/farmacología , Citidina/farmacocinética , Administración Oral , Animales , Área Bajo la Curva , Disponibilidad Biológica , Cromatografía Líquida de Alta Presión , Citidina/administración & dosificación , Metilasas de Modificación del ADN/antagonistas & inhibidores , Femenino , Infusiones Intravenosas , Macaca mulatta , Masculino , Ratones , Ratas
14.
J Med Chem ; 48(21): 6549-52, 2005 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-16220971

RESUMEN

Human glutathione (GSH) transferase (hGSTP1-1) catalyzes the conversion of antitumor 2-crotonyloxymethyl-2-cycloalkenones (COMCs) to highly reactive exocyclic enone alkylating agents. In vitro efficacy studies show that the cytotoxicities of the COMCs directly correlate with the level of expression of GSTP1-1 in MCF-7(piGST) versus MCF-7wt breast tumors, indicating that the exocyclic enones are the actual cytotoxic species. The COMCs are a potentially important new class of prodrugs, which can specifically target multi-drug-resistant tumors overexpressing hGSTP1-1.


Asunto(s)
Antineoplásicos Alquilantes/farmacocinética , Gutatión-S-Transferasa pi/biosíntesis , Profármacos/farmacocinética , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacología , Neoplasias de la Mama , Línea Celular Tumoral , Ciclohexanonas/metabolismo , Ciclohexanonas/farmacocinética , Ciclohexanonas/farmacología , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Profármacos/metabolismo , Profármacos/farmacología , Relación Estructura-Actividad
15.
Cancer Chemother Pharmacol ; 55(1): 21-32, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15338192

RESUMEN

PURPOSE: 17-demethoxy 17-[[(2-dimethylamino)ethyl]amino]geldanamycin (17DMAG, NSC 707545) is a water-soluble analogue of 17-(allylamino)-17-demethoxygeldanamycin (17AAG), a compound currently in clinical trials. These preclinical studies: (1) characterized 17DMAG concentrations in plasma, normal tissues, and tumor after i.v. delivery to mice; and (2) correlated tumor and normal tissue 17DMAG concentrations with alterations in heat shock protein 90 (HSP90) and selected HSP90-chaperoned proteins. METHODS: At specified times after i.v. administration of 75 mg/kg 17DMAG, SCID mice bearing s.c. MDA-MB-231 human breast xenografts were killed and plasma and tissues were retained. 17DMAG concentrations were determined by HPLC. Raf-1, heat shock protein 70 (HSP70), and HSP90 in tissues were determined by Western blotting. RESULTS: Peak plasma 17DMAG concentration was 15.4+/-1.4 microg/ml. The area under the plasma 17DMAG concentration versus time curve was 1072 microg/ml min, corresponding to a total body clearance of 70 ml/kg/min. Peak 17DMAG concentrations in liver (118.8+/-5.7 microg/g), kidney (122.9+/-10.6 microg/g), heart (81.3+/-8.1 microg/g), and lung (110.6+/-25.4 microg/g) occurred at 5-10 min, while peak concentrations in spleen (70.6+/-9.6 microg/g) and tumor (9.0+/-1.0 microg/g) occurred at 30-45 min. At 48 h, 17DMAG was detectable in tumor but not in any normal tissue. Raf-1 in tumors of 17DMAG-treated mice killed at 4, 7, 24 and 48 h was about 20% lower than in tumors from vehicle-treated mice. HSP90 and HSP70 in tumors of 17DMAG-treated animals were significantly lower than in tumors of control animals at 4, 7, and 24 h. Hepatic Raf-1 was decreased by more than 60% at all times after 17DMAG treatment; however, hepatic HSP90 was not affected. HSP70 was undetectable in livers of vehicle-treated mice or mice killed at 2 or 4 h after 17DMAG treatment, but was detected in livers at 7, 24 and 48 h. 17DMAG did not affect renal Raf-1. In contrast, renal HSP70 and HSP90 were decreased by more than 50% at 2 and 4 h after 17DMAG treatment. Renal HSP70 increased approximately twofold above that in kidneys from vehicle-treated control mice at 7 and 24 h, while HSP90 relative protein concentration was no different from that in controls. CONCLUSIONS: Plasma pharmacokinetics of 17DMAG in tumor-bearing mice were similar to those previously reported in nontumor-bearing mice. 17DMAG was distributed widely to tissues but was retained for longer in tumors than normal tissues. Raf-1, HSP90, and HSP70 were altered to different degrees in tumors, livers, and kidneys of 17DMAG-treated animals. These data illustrate the complex nature of the biological responses to 17DMAG.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Quinonas/farmacología , Quinonas/farmacocinética , Animales , Benzoquinonas , Neoplasias de la Mama/patología , Femenino , Infusiones Intravenosas , Lactamas Macrocíclicas , Ratones , Ratones SCID , Quinonas/administración & dosificación , Distribución Tisular , Trasplante Heterólogo
16.
Clin Cancer Res ; 10(19): 6669-76, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15475457

RESUMEN

PURPOSE: In colorectal, breast, and head and neck cancers, response to 5-fluorouracil is associated with low expression of thymidylate synthase. In contrast, tumors with high expression of thymidylate synthase may be more sensitive to prodrugs such as 1-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl) uracil (FAU) that are activated by thymidylate synthase. These studies were designed to evaluate FAU as a potential therapeutic and diagnostic probe. EXPERIMENTAL DESIGN: [18F]-FAU and [3H]-FAU were synthesized with >97% radiochemical purity. [3H]-FAU or [18F]-FAU was administered intravenously to severe combined immunodeficient mice bearing either HT29 (low thymidylate synthase) or LS174T (high thymidylate synthase) human colon cancer xenografts. Four hours after [3H]-FAU dosing, tissue distribution of total radioactivity and incorporation of 1-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl) 5-methyluracil (FMAU), derived from thymidylate synthase activation of FAU, into tumor DNA was measured. Positron emission tomography (PET) images were obtained for 90 minutes after injection of [18F]-FAU. Thymidylate synthase activity was determined in vitro in tumors from untreated mice by [3H] release from [3H]dUMP. Each cell line was incubated in vitro with [3H]-FAU or [3H]-FMAU in the absence or presence of 5-fluoro-2'-deoxyuridine (FdUrd) and then was analyzed for incorporation of radiolabel into DNA. RESULTS: Thymidylate synthase enzymatic activity in LS174T xenografts was approximately 3.5-fold higher than in HT29 xenografts, and incorporation of radioactivity derived from [3H]-FAU into LS174T DNA was approximately 2-fold higher than into HT29 DNA. At 240 minutes, radioactivity derived from [3H]-FAU was approximately 2-fold higher in tumors than in skeletal muscle. At times up to 90 minutes, PET imaging detected only small differences in uptake of [18F]-FAU between the tumor types. Fluorine-18 in skeletal muscle was higher than in tumor for the first 90 minutes and plateaued earlier, whereas [18F] in tumor continued to increase during the 90-minute imaging period. For both cell lines in vitro, FdUrd decreased the rate of incorporation of [3H]-FAU into DNA, whereas the incorporation of [3H]-FMAU was increased. CONCLUSIONS: These results for FAU incorporation into DNA in vitro and in vivo further support clinical evaluation of FAU as a therapeutic agent in tumors with high concentrations of thymidylate synthase that are less likely to respond to 5-fluorouracil treatment. The high circulating concentrations of thymidine reported in mice may limit their utility in evaluating FAU as a PET probe.


Asunto(s)
Arabinofuranosil Uracilo/análogos & derivados , Arabinofuranosil Uracilo/farmacocinética , Neoplasias Colorrectales/tratamiento farmacológico , Fluorouracilo/análogos & derivados , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Arabinofuranosil Uracilo/uso terapéutico , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , ADN de Neoplasias/metabolismo , Femenino , Fluorouracilo/farmacocinética , Fluorouracilo/uso terapéutico , Células HT29 , Humanos , Ratones , Ratones SCID , Tomografía de Emisión de Positrones , Timidilato Sintasa/metabolismo , Factores de Tiempo , Distribución Tisular , Tritio
17.
Clin Cancer Res ; 8(9): 2992-9, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12231546

RESUMEN

One possible explanation for variable tumor response within a single patient may be related to delivery of chemotherapeutic agents to the tumors. Microdialysis was used to evaluate inter- and intratumoral disposition of unbound platinum (Pt) after cisplatin administration to mice bearing B16 murine melanoma tumors or H23 human NSCLC xenografts. Before i.v. dosing with cisplatin (3 or 10 mg/kg), microdialysis probes were placed into the right and left sides of each tumor, and serial extracellular fluid (ECF) samples were collected for 2 h. After microdialysis, tumor samples were obtained at each probe site to measure total Pt and Pt-DNA adducts. In a separate study, serial plasma samples (n = 3 mice/time point) were obtained between 5 min and 2 h. Unbound Pt in tumor ECF and plasma and total Pt in tumor homogenates were measured by flameless atomic absorption spectrophotometry. Pt-DNA adducts in tumor samples were measured via (32)P-postlabeling. Area under the plasma (AUC(P)) and tumor ECF (AUC(ECF)) concentration-time curves of unbound Pt were calculated. Factor VIII expression was measured by immunohistochemistry in tumor samples. After administration of 3 or 10 mg/kg of cisplatin to mice bearing B16 tumors, there was a proportional increase in AUC(PL) with dose; however, there was not a proportional increase in AUC(ECF). There was a relatively high (30-fold) inter- and low (2.5-fold) intratumoral variability in AUC(ECF). AUC(ECF) correlated better with Pt-DNA adduct formation than did total Pt concentration in tumors. There was no relationship between Factor VIII expression and Pt exposure in tumors. The variable penetration of Pt from plasma into tumor ECF may be associated with variable response of tumors.


Asunto(s)
Antineoplásicos/farmacocinética , Carcinoma de Pulmón de Células no Pequeñas/química , Cisplatino/farmacocinética , Neoplasias Pulmonares/química , Melanoma Experimental/química , Platino (Metal)/análisis , Animales , Antineoplásicos/administración & dosificación , Área Bajo la Curva , Carcinoma de Pulmón de Células no Pequeñas/irrigación sanguínea , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Cisplatino/administración & dosificación , Aductos de ADN , ADN de Neoplasias/efectos de los fármacos , Factor VIII/biosíntesis , Factor VIII/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Microdiálisis , Modelos Biológicos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Neovascularización Patológica/tratamiento farmacológico , Organismos Libres de Patógenos Específicos , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Med Chem ; 46(1): 194-6, 2003 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-12502374

RESUMEN

The antitumor activity of 2-crotonyloxymethyl-2-cyclohexenone (COMC-6) is not the result of the GSH conjugate (GSMC-6) formed inside tumor cells, as the diethyl ester prodrug form of GSMC-6 displays little antitumor activity with B16 melanotic melanoma in vitro (IC(50) > 460 microM) versus COMC-6 (IC(50) 0.041 microM) and its five- and seven-membered ring homologues. Antitumor activity probably results from a reactive intermediate that forms during conjugation of the COMCs with intracellular GSH.


Asunto(s)
Antineoplásicos/farmacología , Crotonatos/farmacología , Ciclohexanonas/farmacología , Cicloparafinas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , División Celular/efectos de los fármacos , Crotonatos/síntesis química , Crotonatos/química , Ciclohexanos/síntesis química , Ciclohexanos/química , Ciclohexanos/farmacología , Ciclohexanonas/síntesis química , Ciclohexanonas/química , Cicloparafinas/síntesis química , Cicloparafinas/química , Glutatión/química , Glutatión/metabolismo , Ratones , Relación Estructura-Actividad , Células Tumorales Cultivadas
19.
Cancer Chemother Pharmacol ; 53(4): 329-36, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14673619

RESUMEN

PURPOSE: SPI-077 and SPI-077 B103 are formulations of cisplatin encapsulated in pegylated STEALTH liposomes that accumulate in tumors. However, the extent to which active platinum (Pt) is released from the liposome is unknown. Thus, we evaluated the disposition of encapsulated and released Pt in plasma and tumors after administration of STEALTH liposomal and nonliposomal cisplatin. METHODS: Cisplatin (10 mg/kg), SPI-077 (10 mg/kg), and SPI-077 B103 (5 mg/kg) were administered i.v. to mice bearing B16 murine melanoma tumors. Microdialysis probes were placed into the right and left sides of each tumor, and serial samples were collected from tumor extracellular fluid (ECF) after administration of each agent. After each microdialysis procedure, tumor samples were obtained at each probe site to measure total Pt and Pt-DNA adducts. In a separate study, serial plasma samples (three mice per time point) were obtained. Unbound Pt in tumor ECF and plasma, and total Pt in tumor homogenates were measured by flameless atomic absorption spectrophotometry. Area under the tumor ECF (AUC(ECF)) concentration versus time curves of unbound Pt were calculated. Intrastrand GG (Pt-GG) and AG (Pt-AG) Pt-DNA adducts were measured via (32)P-postlabeling. RESULTS: Mean+/-SD peak concentrations of total Pt in tumor homogenates after administration of cisplatin, SPI-077, and SPI-077 B-103 were 3.2+/-1.9, 11.9+/-3.0, and 3.5+/-0.3 microg/g, respectively. After cisplatin, mean+/-SD AUC(ECF) of unbound Pt was 0.72+/-0.46 microg/ml.h. There was no detectable unbound Pt in tumor ECF after SPI-077 or SPI-077 B-103 treatment. Mean+/-SD peak concentration of Pt-GG DNA adducts after administration of cisplatin, SPI-077, and SPI-077 B-103 were 13.1+/-3.3, 3.5+/-1.3, and 2.1+/-0.3 fmol Pt/microg DNA, respectively. CONCLUSION: This study suggests that more SPI-077 and SPI-077 B103 distribute into tumors, but release less Pt into tumor ECF, and form fewer Pt-DNA adducts than does cisplatin.


Asunto(s)
Antineoplásicos/farmacocinética , Cisplatino/metabolismo , Cisplatino/farmacocinética , Aductos de ADN/metabolismo , ADN de Neoplasias/metabolismo , Melanoma Experimental/metabolismo , Animales , Antineoplásicos/administración & dosificación , Área Bajo la Curva , Cisplatino/administración & dosificación , Femenino , Inyecciones Intravenosas , Ratones , Ratones Endogámicos C57BL , Microdiálisis , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Metabolism ; 60(10): 1465-74, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21489580

RESUMEN

Inositol hexaphosphate (IP(6)) is effective in preclinical cancer prevention and chemotherapy. In addition to cancer, IP(6) has many other beneficial effects for human health, such as reduction in risk of developing cardiovascular disease and diabetes and inhibition of kidney stone formation. Studies presented here describe the pharmacokinetics, tissue distribution, and metabolism of IP(6) following intravenous (IV) or per os (PO) administration to mice. SCID mice bearing MDA-MB-231 xenografts were treated with 20 mg/kg IP(6) (3 µCi per mouse [(14)C]-uniformly ring-labeled IP(6)) and euthanized at various times after IP(6) treatment. Plasma and tissues were analyzed for [(14)C]-IP(6) and metabolites by high-performance liquid chromatography with radioactivity detection. Following IV administration of IP(6), plasma IP(6) concentrations peaked at 5 minutes and were detectable until 45 minutes. Liver IP(6) concentrations were more than 10-fold higher than plasma concentrations, whereas other normal tissue concentrations were similar to plasma. Only inositol was detected in xenografts. After PO administration, IP(6) was detected in liver; but only inositol was detectable in other tissues. After both IV and PO administration, exogenous IP(6) was rapidly dephosphorylated to inositol; however, alterations in endogenous IPs were not examined.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Ácido Fítico/farmacocinética , Animales , Neoplasias de la Mama/patología , Carcinoma/patología , Línea Celular Tumoral , Femenino , Humanos , Infusiones Intravenosas , Dosis Máxima Tolerada , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Trasplante de Neoplasias , Ácido Fítico/administración & dosificación , Distribución Tisular , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA