Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Chemother ; 32(6): 310-322, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32441565

RESUMEN

We studied the predictive value for response and toxicity of functional polymorphisms in genes involved in the oxaliplatin/fluorouracil pathway in colorectal cancer patients. One hundred and twenty-seven (127) patients were treated with curative intended surgery followed by adjuvant chemotherapy with FOLFOX (fluorouracil, leucovorin and oxaliplatin) regimen. The median age was 65.53 (27-80) years (66.9% male, 59.1% rectum). The median follow-up was 8.5 years (IQR, 4.1-9.4). At the end of follow-up, 59 patients (46.5%) had relapsed or died in the whole study population. We did find that XRCC1GG genotype is associated with a higher risk of developing haematologic toxicity. Furthermore, we report a significant association of the TS 3'UTR 6 bp/6 bp polymorphism and the XRCC1 rs25487 with a higher risk of developing anaemia and diarrhoea, respectively. On the other hand, none of the studied polymorphisms showed clinically relevant association with disease-free survival and overall survival or early failure to adjuvant FOLFOX therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Quimioterapia Adyuvante , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/cirugía , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endonucleasas/genética , Endonucleasas/metabolismo , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/efectos adversos , Fluorouracilo/farmacocinética , Humanos , Leucovorina/administración & dosificación , Leucovorina/efectos adversos , Leucovorina/farmacocinética , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Compuestos Organoplatinos/administración & dosificación , Compuestos Organoplatinos/efectos adversos , Compuestos Organoplatinos/farmacocinética , Oxaliplatino/administración & dosificación , Oxaliplatino/efectos adversos , Oxaliplatino/farmacocinética , Polimorfismo Genético , Regiones Promotoras Genéticas , Timidilato Sintasa/genética , Timidilato Sintasa/metabolismo
2.
Eur J Pharm Sci ; 119: 22-30, 2018 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-29626594

RESUMEN

BACKGROUND: The evaluation of the efficacy and toxicity of hyperthermic intraoperative peritoneal chemotherapy presents some difficulties, due in part to the lack of information about the pharmacokinetic behavior of the drugs administered in this procedure. The aim of this study was to characterize the population pharmacokinetics of hyperthermic intraoperative peritoneal oxaliplatin in Wistar rats and to evaluate the effect of treatment-related covariates dose, instillation time and temperature on the pharmacokinetic parameters. METHODS: Oxaliplatin peritoneal and plasma concentrations from 37 rats treated by either intravenous or intraperitoneal oxaliplatin administrations under different instillation times, temperatures and doses were analyzed according to a population pharmacokinetic approach using the software NONMEM V7.3®. RESULTS: Intraperitoneal (n = 115) and plasma (n = 263) concentrations were successfully described according to a two-compartment model with first order absorption. No significant effect of dose, temperature and instillation time on pharmacokinetic parameters was found. However, an abrupt decrease in the elimination process was observed, reflected in the structural pharmacokinetic model through a modification in clearance. The typical parameters values and the interindividual variability (CV %) in clearance, central and peripheral volume of distribution were 3.25 mL/min (39.1%), 53.6 mL (37.8%) and 54.1 mL (77.3%), respectively. Clearance decreased to 0.151 mL/min (39.1%) when the instillation was still ongoing, at 31.4 min. One of the possible reasons behind the clearance decrease would be an alteration of renal function due to surgery and/or hyperthermia. CONCLUSIONS: This study described the deterioration of the drug elimination process due to the procedure, and estimated the time at which this deterioration is most likely to occur. In addition, dose, instillation time and temperature had no influence in the PK parameters.


Asunto(s)
Antineoplásicos/farmacocinética , Hipertermia Inducida , Modelos Biológicos , Compuestos Organoplatinos/farmacocinética , Administración Intravenosa , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/sangre , Terapia Combinada , Inyecciones Intraperitoneales , Masculino , Compuestos Organoplatinos/administración & dosificación , Compuestos Organoplatinos/sangre , Oxaliplatino , Peritoneo/metabolismo , Ratas Wistar
3.
Bull Exp Biol Med ; 163(3): 349-351, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28744651

RESUMEN

Antimetastatic activity of Platin in lyophilized liposomes stored for 7 years after fabrication was evaluated. The main flaw of liposomes as vehicles for drug delivery to the tumors is their high affinity for the liver, which accumulates a great amount thereof. This property of liposomes can be used for adjuvant therapy of operable primary tumors metastasizing to the liver. It is shown on the model of mouse GA-1 tumor metastases in the liver that platinum(II) complex compound Platin in phosphatidylcholine-cholesterol liposomes, stored for 7 years after lyophilization, causes complete cure of 40% animals, while free Platin prolongs the lifespan of mice with tumors by only 31.7% vs. control (no treatment).


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Ehrlich/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Liposomas/administración & dosificación , Neoplasias Hepáticas/tratamiento farmacológico , Compuestos Organoplatinos/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Carcinoma de Ehrlich/metabolismo , Carcinoma de Ehrlich/mortalidad , Carcinoma de Ehrlich/patología , Colesterol/química , Esquema de Medicación , Composición de Medicamentos , Estabilidad de Medicamentos , Femenino , Liofilización , Inyecciones Intravenosas , Liposomas/química , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/secundario , Ratones , Compuestos Organoplatinos/química , Compuestos Organoplatinos/farmacocinética , Fosfatidilcolinas/química , Análisis de Supervivencia
4.
Adv Healthc Mater ; 6(16)2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28504409

RESUMEN

Intratumoral (IT) drug injections reduce systemic toxicity, but delivered volumes and distribution can be inconsistent. To improve IT delivery paradigms, porphyrin-phospholipid (PoP) liposomes are passively loaded with three hydrophilic cargos: sulforhodamine B, a fluorophore; gadolinium-gadopentetic acid, a magnetic resonance (MR) agent; and oxaliplatin, a colorectal cancer chemotherapeutic. Liposome composition is optimized so that cargo is retained in serum and storage, but is released in less than 1 min with exposure to near infrared light. Light-triggered release occurs with PoP-induced photooxidation of unsaturated lipids and all cargos release concurrently. In subcutaneous murine colorectal tumors, drainage of released cargo is delayed when laser treatment occurs 24 h after IT injection, at doses orders of magnitude lower than systemic ones. Delayed light-triggering results in substantial tumor shrinkage relative to controls a week following treatment, although regrowth occurs subsequently. MR imaging reveals that over this time frame, pools of liposomes within the tumor migrate to adjacent regions, possibly leading to altered spatial distribution during triggered drug release. Although further characterization of cargo loading and release is required, this proof-of-principle study suggests that multimodal theranostic IT delivery approaches hold potential to both guide injections and interpret outcomes, in particular when combined with chemo-phototherapy.


Asunto(s)
Antineoplásicos , Medios de Contraste , Colorantes Fluorescentes , Liposomas , Fotoquimioterapia/métodos , Nanomedicina Teranóstica/métodos , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Medios de Contraste/química , Medios de Contraste/farmacocinética , Femenino , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacocinética , Liposomas/química , Liposomas/farmacocinética , Liposomas/farmacología , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos BALB C , Compuestos Organoplatinos/química , Compuestos Organoplatinos/farmacocinética , Compuestos Organoplatinos/farmacología , Oxaliplatino , Fosfolípidos/química , Porfirinas/química
5.
Cancer Chemother Pharmacol ; 79(3): 621-627, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28168311

RESUMEN

PURPOSE: In peritoneal metastasis condition, the fact that most of the disease is limited to the peritoneal cavity laid the foundations for a surgical treatment, including intraperitoneal hyperthermic chemotherapy (HIPEC). The aim of this study was to evaluate the impact of the surgical procedures implied in open HIPEC technique, referred to laparotomy procedures followed by an intraperitoneal hyperthermic instillation (LIHI) on oxaliplatin tissue distribution and elimination. To delimit the influence of this procedure alone, oxaliplatin was administered as an intravenous (iv) bolus in both groups. METHODS: An experimental model in Wistar rats was employed, and LIHI was evaluated as a dichotomous covariate by using a population pharmacokinetic (PK) approach. Rats were randomized in two groups receiving 1.5 mg iv oxaliplatin alone or 1.5 mg iv oxaliplatin under LIHI conditions, carrying out a hyperthermic 5% dextrose instillation. The oxaliplatin plasma concentrations were characterized by an open two-compartment PK model. RESULTS: Results concluded that surgical conditions affect the oxaliplatin elimination and distribution from blood to peripheral tissues, increasing the systemic drug exposure. Concretely, oxaliplatin peripheral volume of distribution, and clearance decreased by 48.6% and 55.3%, respectively, compared to the control group that resulted in a two-fold increase of the area under the concentration time curve. CONCLUSIONS: Comparison in clinical practice of oxaliplatin PK parameters obtained after iv administrations with those obtained after HIPEC interventions must be done carefully. This would limit the use of iv PK parameters to simulate new scenarios for oxaliplatin in HIPEC.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Hipertermia Inducida , Laparotomía/métodos , Compuestos Organoplatinos/administración & dosificación , Compuestos Organoplatinos/farmacocinética , Administración Intravenosa , Animales , Área Bajo la Curva , Inyecciones Intraperitoneales , Masculino , Modelos Estadísticos , Oxaliplatino , Neoplasias Peritoneales/tratamiento farmacológico , Ratas , Ratas Wistar , Reproducibilidad de los Resultados , Distribución Tisular
6.
Anticancer Drugs ; 28(3): 281-288, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27997436

RESUMEN

Dihydromyricetin (DMY), extracted from the Chinese herbal medicine Ampelopsis grossedentata, possesses antitumor potential in different types of human cancer cells. Hence, its effects on drug resistance and molecular mechanisms in colorectal cancer (CRC) are still unknown. In our present study, we observed that DMY enhanced the chemosensitivity to oxaliplatin (OXA). DMY increased OXA-induced apoptosis and reduced 5(6)-carboxy-2',7'-dichlorofluorescein accumulation in OXA-resistant CRC HCT116/L-OHP cells. Our mechanistic study suggested that DMY treatment inhibited multidrug resistance protein 2 (MRP2) expression levels and promoter activity, indicating that DMY reduced not only MRP2 transcriptional and translational levels but also its function. Additional experiments indicated that the nuclear translocation of nuclear factor-erythroid 2 p45 related factor 2, a MRP2 regulator, was also inhibited by DMY. In summary, our study provided the first direct evidence that the inhibitory effects of DMY on MRP2 expression in OXA-resistant CRC cells were closely associated with the inhibition of nuclear factor-erythroid 2 p45 related factor 2 signaling. DMY could be a potential candidate for CRC chemotherapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Flavonoles/farmacología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Compuestos Organoplatinos/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Neoplasias Colorrectales/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Flavonoles/administración & dosificación , Células HCT116 , Humanos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/biosíntesis , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas de Neoplasias/metabolismo , Compuestos Organoplatinos/administración & dosificación , Compuestos Organoplatinos/farmacocinética , Oxaliplatino , Regiones Promotoras Genéticas
7.
Clin Pharmacokinet ; 56(2): 165-177, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27393140

RESUMEN

The combination of hepatic artery infusion (HAI) of irinotecan, 5-fluorouracil and oxaliplatin with intravenous cetuximab has safely achieved prolonged survival in colorectal cancer patients with extensive liver metastases and prior treatment. Systemic exposure to the drugs or their main metabolites was determined during the first course of chronomodulated triplet HAI in 11 patients and related to toxicities after one or three courses. Consistent trends were found between the area under the plasma concentration-time curve (AUC) values of irinotecan, 7-ethyl-10-hydroxycamptothecin (SN38; a bioactive metabolite), total oxaliplatin and platinum ultrafiltrate (P-UF), on the one hand, and subsequent leukopenia severity, on the other hand. Moreover, the maximum plasma concentration (C max) and the AUC of P-UF significantly predicted grades of diarrhoea (p = 0.004 and 0.017, respectively) and anaemia (p = 0.001 and 0.008, respectively) after the first course. Systemic drug exposure helps explain both the adverse events and the low rate of extrahepatic progression-a usual drawback of HAI chemotherapy-thus supporting upfront testing of the regimen. Systems optimization of chronomodulated HAI delivery could further reduce adverse events.


Asunto(s)
Camptotecina/análogos & derivados , Cronoterapia de Medicamentos , Fluorouracilo/farmacocinética , Arteria Hepática/metabolismo , Neoplasias Hepáticas/sangre , Compuestos Organoplatinos/farmacocinética , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica , Camptotecina/administración & dosificación , Camptotecina/farmacocinética , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/tratamiento farmacológico , Europa (Continente)/epidemiología , Femenino , Fluorouracilo/administración & dosificación , Arteria Hepática/efectos de los fármacos , Humanos , Infusiones Intraarteriales/métodos , Irinotecán , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/secundario , Masculino , Persona de Mediana Edad , Compuestos Organoplatinos/administración & dosificación , Oxaliplatino
8.
Dokl Biochem Biophys ; 467(1): 92-4, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27193706

RESUMEN

The article presents the results of investigation of antitumor properties of platinum-arabinogalactan complex. We showed the ability of the complex to inhibit the growth of Ehrlich ascites tumor cells. It is found that the distribution of the platinum-arabinogalactan complex is not specific only for tumor cells in mice. The complex was found in all tissues and organs examined (ascites cells, embryonic cells, kidney, and liver). The mechanism of action of the arabinogalactan-platinum complex may be similar to cisplatin as the complex is able to accumulate in tumor cells.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Ehrlich/tratamiento farmacológico , Galactanos/farmacología , Compuestos Organoplatinos/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Ascitis/tratamiento farmacológico , Ascitis/metabolismo , Carcinoma de Ehrlich/metabolismo , Cisplatino/farmacología , Evaluación Preclínica de Medicamentos , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Galactanos/síntesis química , Galactanos/farmacocinética , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Masculino , Ratones Endogámicos ICR , Microscopía Fluorescente , Trasplante de Neoplasias , Compuestos Organoplatinos/síntesis química , Compuestos Organoplatinos/farmacocinética
9.
PLoS One ; 11(2): e0148739, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26859833

RESUMEN

BACKGROUND: Adjuvant chemotherapy for colorectal cancer is mainly based on the combination of 5-fluorouracil, folinic acid and oxaliplatin (FOLFOX-4). The pharmacological target of oxaliplatin remains intracellular and therefore dependent on its entry into cells. The intracellular distribution of oxaliplatin is mediated by organic cation transporters 1, 2 and 3 (OCT1, 2 and 3), copper transporter 1 (CTR1) and ATPase Cu2+ transporting beta polypeptide (ATP7B) and may modulate the efficacy of oxaliplatin-based chemotherapy. The aim of this study was to perform a retrospective study to assess the relation between the expression of oxaliplatin transporters in colorectal cancer before chemotherapy and the response to FOLFOX-4 adjuvant chemotherapy in responder and non-responder patients. METHODS: This retrospective study was conducted at a single center (University Hospital of Clermont-Ferrand, France). The target population was patients with resectable colorectal cancer operated between 2006 and 2013. Inclusion criteria were defined for the responder patients as no cancer recurrence 3 years after the end of chemotherapy, and for the non-responder patients as cancer recurrence within 1 year. Other inclusion criteria were stages IIb-IV cancers, first-line adjuvant FOLFOX-4 chemotherapy, and the availability of resected primary tumor samples. Exclusion criteria were preoperative chemotherapy and/or radiotherapy, a targeted therapy, other anticancer drugs, cancer recurrence between the first and the third year after the end of chemotherapy and follow-up < 3 years. Immunostaining of oxaliplatin transporters (OCT1, 2, 3, CTR1 and ATP7B) and Ki-67 was assessed in tumor samples. RESULTS: Retrospectively, 31 patients have been selected according to inclusion and exclusion criteria (15 responders and 16 non-responders). Before FOLFOX-4 regimen, OCT3 expression was significantly lower in responder patients compared to non-responders (p<0.001). According to multivariate analysis, OCT3 remains an independent criterion for adjuvant FOLFOX chemotherapy response (p = 0.039). No significant relation is reported between chemotherapy response and the expression of OCT1 (p = 0.49), OCT2 (p = 0.09), CTR1 (p = 0.45), ATP7B (p = 0.94) and Ki-67 (p = 0.34) in tumors. CONCLUSIONS: High expression of OCT3 could be an independent factor related to resistance to FOLFOX-4 chemotherapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proteínas de Transporte de Catión/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Compuestos Organoplatinos/metabolismo , Adenosina Trifosfatasas/metabolismo , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Quimioterapia Adyuvante , Transportador de Cobre 1 , ATPasas Transportadoras de Cobre , Femenino , Fluorouracilo/uso terapéutico , Humanos , Inmunohistoquímica , Leucovorina/uso terapéutico , Masculino , Persona de Mediana Edad , Proteínas de Transporte de Catión Orgánico/metabolismo , Transportador 1 de Catión Orgánico/metabolismo , Transportador 2 de Cátion Orgánico , Compuestos Organoplatinos/farmacocinética , Compuestos Organoplatinos/uso terapéutico , Oxaliplatino , Estudios Retrospectivos , Resultado del Tratamiento
10.
Cancer Chemother Pharmacol ; 74(3): 571-82, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25053386

RESUMEN

PURPOSE: First, to evaluate the peritoneal (IP), plasma ultrafiltrated (UF) and protein-bound (B) pharmacokinetics (PK) of oxaliplatin after intraperitoneal hyperthermic chemoperfusion (HIPEC) following cytoreductive surgery. Second, to evaluate the relationship between oxaliplatin exposure and observed toxicity. METHODS: IP, UF, and B concentrations from 75 patients treated by 30-min oxaliplatin-based HIPEC procedures were analysed according to a pharmacokinetic modelling approach using NONMEM. Oxaliplatin was administered in a 5 % dextrose solution (2 L/m(2)) at 360 (n = 58) or 460 mg/m(2) (n = 17). The most frequently observed toxicities were related to the peritoneal, systemic exposures and to the parameters corresponding to the oxaliplatin absorption from peritoneal cavity into plasma. RESULTS: IP (n = 536), UF (n = 669) and B (n = 661) concentrations were simultaneously described according to a five-compartment PK model with irreversible nonlinear binding from UF to B according to a Michaelis-Menten equation. The mean (±SD) maximum fraction of dose absorbed and elimination half-life from the peritoneum was 53.7 % (±8.5) and 0.49 h (±0.1), respectively. The mean (±SD) ratio AUC(IP)/AUC(UF) was 5.3 (±2) confirming the pharmacokinetic advantage of the procedure. Haemoperitoneum (22.7 %), neuropathy (18.7 %), grade 3/4 thrombocytopenia (13.3 %) were the most frequently reported toxicities. AUC(UF) accounts for approximately 12 % of the variation in the maximum percentage of platelet decrease (r = 0.35, p = 0.002). Thrombocytopenia was correlated with higher AUCUF, partly dependent on the extent and rate of oxaliplatin absorption. CONCLUSIONS: Despite a common dose administered, variability in peritoneal and systemic oxaliplatin exposures are observed, leading to differences in haematological toxicity between patients.


Asunto(s)
Quimioterapia del Cáncer por Perfusión Regional/efectos adversos , Compuestos Organoplatinos/administración & dosificación , Compuestos Organoplatinos/efectos adversos , Compuestos Organoplatinos/farmacocinética , Neoplasias Peritoneales/tratamiento farmacológico , Trombocitopenia/inducido químicamente , Adulto , Anciano , Área Bajo la Curva , Semivida , Humanos , Hipertermia Inducida/efectos adversos , Infusiones Parenterales/efectos adversos , Persona de Mediana Edad , Modelos Teóricos , Compuestos Organoplatinos/sangre , Oxaliplatino , Neoplasias Peritoneales/patología , Ultrafiltración
11.
Invest Clin ; 55(2): 185-202, 2014 Jun.
Artículo en Español | MEDLINE | ID: mdl-24974634

RESUMEN

Gastric cancer (GC) is often diagnosed at later stages due to the lack of specificity of symptoms associated with the neoplasm, causing high mortality rates worldwide. The first line of adjuvant and neoadjuvant treatment includes cytotoxic fluoropyrimidines and platin-containing compounds which cause the formation of DNA adducts. The clinical outcome with these antineoplastic agents depends mainly on tumor sensitivity, which is conditioned by the expression level of the drug targets and the DNA-repair system enzymes. In addition, some germ line polymorphisms, in genes linked to drug metabolism and response to chemotherapy, have been associated with poor responses and the development of adverse effects, even with fatal outcomes in GC patients. The identification of genomic biomarkers, such as individual gene polymorphisms or differential expression patterns of specific genes, in a patient-by-patient context with potential clinical application is the main focus of current pharmacogenomic research, which aims at developing a rational and personalized therapy (i.e., a therapy that ensures maximum efficacy with no predictable side effects). However, because of the future application of genomic technologies in the clinical setting, it is necessary to establish the prognostic value of these genomic biomarkers with genotype-phenotype association studies and to evaluate their prevalence in the population under treatment. These issues are important for their cost-effectiveness evaluation, which determines the feasibility of using these medical genomic research products for GC treatment in the clinical setting.


Asunto(s)
Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Gástricas/tratamiento farmacológico , Antineoplásicos/efectos adversos , Antineoplásicos/clasificación , Transporte Biológico/genética , Biomarcadores , Biotransformación/genética , Capecitabina , Terapia Combinada , Desoxicitidina/efectos adversos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacocinética , Desoxicitidina/uso terapéutico , Combinación de Medicamentos , Resistencia a Antineoplásicos/genética , Enzimas/genética , Etnicidad/genética , Fluorouracilo/efectos adversos , Fluorouracilo/análogos & derivados , Fluorouracilo/farmacocinética , Fluorouracilo/uso terapéutico , Gastrectomía , Humanos , México , Terapia Molecular Dirigida , Compuestos Organoplatinos/farmacocinética , Ácido Oxónico/farmacocinética , Selección de Paciente , Farmacogenética , Medicina de Precisión , Profármacos/farmacocinética , Neoplasias Gástricas/genética , Neoplasias Gástricas/cirugía , Tegafur/farmacocinética
12.
Invest. clín ; Invest. clín;55(2): 185-202, jun. 2014. ilus, tab
Artículo en Español | LILACS | ID: lil-749976

RESUMEN

Debido a la inespecificidad de los síntomas, el cáncer gástrico (CG) es diagnosticado frecuentemente en etapas avanzadas, lo que da cuenta de los altos índices de mortalidad debido a esta neoplasia a nivel mundial. El esquema de tratamiento adyuvante o neoadyuvante en los países occidentales incluye el uso de fluoropirimidinas citotóxicas y compuestos de platino formadores de aductos en el ADN. La respuesta clínica al tratamiento con estos fármacos depende principalmente de la sensibilidad del tumor, la cual a su vez está condicionada por el nivel de expresión de los blancos terapéuticos y de las enzimas de reparación del ADN. Sumado a esto, algunos polimorfismos de línea germinal en genes asociados al metabolismo y a la respuesta a estos fármacos, han mostrado asociación con respuestas pobres y con el desarrollo de eventos adversos, incluso con resultados fatales. La identificación de biomarcadores genómicos, en la forma de polimorfismos genéticos o la expresión diferencial de genes específicos asociados a la respuesta quimioterapeútica ha sido motivo de intensa investigación como base para la aplicación de la farmacogenómica en el establecimiento de una terapia farmacológica racional y personalizada del CG. Sin embargo, ante la eventual aplicación de la farmacogenómica en el ámbito clínico, es necesario establecer el valor pronóstico real de dichos biomarcadores mediante los estudios de asociación genotipo-fenotipo, así como su prevalencia en el contexto de cada población de pacientes. Estos aspectos son indispensables al evaluar la relación costo-efectividad de la introducción de los productos de la medicina genómica predictiva en el tratamiento del CG.


Gastric cancer (GC) is often diagnosed at later stages due to the lack of specificity of symptoms associated with the neoplasm, causing high mortality rates worldwide. The first line of adjuvant and neoadjuvant treatment includes cytotoxic fluoropyrimidines and platin-containing compounds which cause the formation of DNA adducts. The clinical outcome with these antineoplastic agents depends mainly on tumor sensitivity, which is conditioned by the expression level of the drug targets and the DNA-repair system enzymes. In addition, some germ line polymorphisms, in genes linked to drug metabolism and response to chemotherapy, have been associated with poor responses and the development of adverse effects, even with fatal outcomes in GC patients. The identification of genomic biomarkers, such as individual gene polymorphisms or differential expression patterns of specific genes, in a patient-by-patient context with potential clinical application is the main focus of current pharmacogenomic research, which aims at developing a rational and personalized therapy (i.e., a therapy that ensures maximum efficacy with no predictable side effects). However, because of the future application of genomic technologies in the clinical setting, it is necessary to establish the prognostic value of these genomic biomarkers with genotype-phenotype association studies and to evaluate their prevalence in the population under treatment. These issues are important for their cost-effectiveness evaluation, which determines the feasibility of using these medical genomic research products for GC treatment in the clinical setting.


Asunto(s)
Humanos , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Gástricas/tratamiento farmacológico , Antineoplásicos/efectos adversos , Antineoplásicos/clasificación , Biomarcadores , Transporte Biológico/genética , Biotransformación/genética , Terapia Combinada , Combinación de Medicamentos , Desoxicitidina/efectos adversos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacocinética , Desoxicitidina/uso terapéutico , Resistencia a Antineoplásicos/genética , Enzimas/genética , Etnicidad/genética , Fluorouracilo/efectos adversos , Fluorouracilo/análogos & derivados , Fluorouracilo/farmacocinética , Fluorouracilo/uso terapéutico , Gastrectomía , México , Terapia Molecular Dirigida , Compuestos Organoplatinos/farmacocinética , Ácido Oxónico/farmacocinética , Selección de Paciente , Farmacogenética , Medicina de Precisión , Profármacos/farmacocinética , Neoplasias Gástricas/genética , Neoplasias Gástricas/cirugía , Tegafur/farmacocinética
13.
Cell Death Dis ; 5: e1014, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24457958

RESUMEN

The higher and selective cytotoxicity of [Pt(O,O'-acac)(γ-acac)(DMS)] toward cancer cell in both immortalized cell lines and in breast cancer cells in primary cultures, stimulated a pre-clinical study so as to evaluate its therapeutic potential in vivo. The efficacy of [Pt(O,O'-acac)(γ-acac)(DMS)] was assessed using a xenograft model of breast cancer developed by injection of MCF-7 cells in the flank of BALB/c nude mice. Treatment of solid tumor-bearing mice with [Pt(O,O'-acac)(γ-acac)(DMS)] induced up to 50% reduction of tumor mass compared with an average 10% inhibition recorded in cisplatin-treated animals. Thus, chemotherapy with [Pt(O,O'-acac)(γ-acac)(DMS)] was much more effective than cisplatin. We also demonstrated enhanced in vivo pharmacokinetics, biodistribution and tolerability of [Pt(O,O'-acac)(γ-acac)(DMS)] when compared with cisplatin administered in Wistar rats. Pharmacokinetics studies with [Pt(O,O'-acac)(γ-acac)(DMS)] revealed prolonged Pt persistence in systemic blood circulation and decreased nefrotoxicity and hepatotoxicity, major target sites of cisplatin toxicity. Overall, [Pt(O,O'-acac)(γ-acac)(DMS)] turned out to be extremely promising in terms of greater in vivo anticancer activity, reduced nephrotoxicity and acute toxicity compared with cisplatin.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Neoplasias de la Mama/tratamiento farmacológico , Compuestos Organoplatinos/administración & dosificación , Animales , Antineoplásicos/efectos adversos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Compuestos Organoplatinos/efectos adversos , Compuestos Organoplatinos/farmacocinética , Ratas , Ratas Wistar , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Gan To Kagaku Ryoho ; 41(12): 2101-3, 2014 Nov.
Artículo en Japonés | MEDLINE | ID: mdl-25731436

RESUMEN

The effects of warming or emulsification with the water-soluble contrast medium, Iomeron (IOM), on reducing the viscosity of miriplatin-lipiodol (MPT-LPD) suspension were studied. Reduction in the viscosity of MPT-LPD suspension was ob- served upon increasing the temperature. Although the O/W MPT-LPD emulsion with a low ratio of MPT-LPD to IOM reduced the viscosity, the effect was lesser than that achieved with the warming treatment. Radiographic images of the liver obtained after administration of the emulsion into the rat portal vein showed that warming resulted in improved intrahepatic distribution of the formulation, which was dependent on the reduction of viscosity. Emulsification also led to better intrahepatic distribution, but this distribution did not depend on the viscosity of the formulation. The MPT-LPD emulsion showed different distribution properties from the MPT-LPD suspension, and it was difficult to estimate the intrahepatic distribution property from the viscosity of the emulsion. Thus, we suggest that emulsification and warming of MPT-LPD are effective methods for improving the intrahepatic distribution of the MPT formulation.


Asunto(s)
Aceite Etiodizado/farmacocinética , Hígado/metabolismo , Compuestos Organoplatinos/farmacocinética , Animales , Emulsiones , Aceite Etiodizado/administración & dosificación , Femenino , Infusiones Intravenosas , Compuestos Organoplatinos/administración & dosificación , Ratas , Ratas Sprague-Dawley , Suspensiones , Viscosidad
15.
Clin Pharmacokinet ; 52(12): 1111-25, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23828617

RESUMEN

BACKGROUND AND OBJECTIVE: Peritoneal carcinomatosis is an abdominal metastatic manifestation of a life-threatening tumour progression requiring standard palliative surgery and/or chemotherapy treatment. The aim of this study was to characterize the immediate neutrophilia response induced by cytoreductive surgery (CRS) and the myelosuppression effect of hyperthermic intraperitoneal oxaliplatin (HIO) in peritoneal carcinomatosis patients. METHODS: Absolute neutrophil counts (ANCs) from 45 patients treated with CRS and HIO diluted in isotonic 4 % icodextrin (cohort A), 21 patients undergoing CRS followed by HIO diluted in isotonic 5 % dextrose (cohort B) and 18 patients treated with CRS without HIO (cohort C) were used to estimate the system-related parameters [baseline ANC (Circ0), mean transit time (MTT) and feedback on proliferation (γ)] and drug-specific (α) parameters of a modified Friberg's model that accounts for the surgical stress-induced neutrophilia. The plasma oxaliplatin concentrations, C(p), were assumed to reduce the proliferation rate of the progenitor cells according to the function α × C(p). Model evaluation and simulations were undertaken to evaluate the effect of the dose, treatment duration and carrier solution on the incidence of severe neutropenia. RESULTS: The typical values [between-subject variability, expressed in coefficient of variation values (%)] of the Circ0, MTT, γ and α were estimated to be 3.58 × 109 cells/L (41.2 %), 144 h (70.9 %), 0.155 and 0.066 L/mg (134.9 %), respectively. Surgical stress induced a maximal 3.37-fold increase in the proliferation rate that was attenuated with a half-life of 10 days, and a maximal 68 % reduction in the MTT that was attenuated with a half-life of 28 days. Age, body surface area, sex, total proteins and carrier solution did not impact the model parameters. The model evaluation evidenced an accurate prediction of the incidence of neutropenia grade ≥2 and/or ≥3. Simulations indicated that (i) the neutropenia was reversible and short-lasting; and (ii) the HIO dose and treatment duration were the main determinants of the severity and duration of neutropenia. CONCLUSION: The time course of neutropenia was well characterized by the model that was developed, which simultaneously accounts for the acute-immediate neutrophilia response induced by CRS and the HIO myelosuppressive effect produced in the bone marrow. This model suggests that higher doses than those evaluated to date could be used in peritoneal carcinomatosis patients without substantially increasing the risk of severe neutropenia.


Asunto(s)
Antineoplásicos/farmacocinética , Modelos Biológicos , Neutrófilos/efectos de los fármacos , Compuestos Organoplatinos/farmacocinética , Neoplasias Peritoneales/metabolismo , Antineoplásicos/administración & dosificación , Terapia Combinada , Procedimientos Quirúrgicos del Sistema Digestivo , Humanos , Hipertermia Inducida , Recuento de Leucocitos , Neutropenia/inducido químicamente , Neutrófilos/citología , Compuestos Organoplatinos/administración & dosificación , Oxaliplatino , Neoplasias Peritoneales/tratamiento farmacológico , Neoplasias Peritoneales/cirugía
16.
Expert Opin Drug Metab Toxicol ; 9(10): 1381-90, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23829480

RESUMEN

INTRODUCTION: Picoplatin was developed as platinum coordination complex to overcome development of resistance, through conjugation to thioles, by the introduction of a methyl-pyridine moiety into the cisplatin parent structure. Pharmacokinetic parameters of the drug, after intravenous and oral application, were studied in solid tumors and clinical Phase I - III trials performed, in particular in NSCLC and small cell lung cancer (SCLC). Results showed low clinical activity of picoplatin. AREAS COVERED: This article presents an overview of the pharmacokinetic assessments of picoplatin in lung cancer. Specifically, the authors address the relationship between disposition and clinical activity of the drug. EXPERT OPINION: Picoplatin failed to overcome resistance to platinum compounds in lung cancer to achieve significant improved survival of most patients. Even highest doses of the drug reaching 150 m/m² given intravenously every 3 weeks were not sufficient to achieve better response than existing chemotherapeutics and the oral bioavailability of a dose of 200 - 400 mg corresponded only to 80 mg/m² iv. Picoplatin therefore seem to be quite ineffective. Picoplatin is expected to overcome tumor resistance in cases which overexpress thiol-conjugating pathways; however, this was not proved in clinical trials. To conclude, this blocked platinum complex is not able to reverse cisplatin resistance to a significant extent in vivo and its mechanisms and kinetics and of DNA damage failed to produce significant clinical results compared to second-line standard therapy for lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Compuestos Organoplatinos/farmacocinética , Administración Oral , Animales , Disponibilidad Biológica , Línea Celular Tumoral , Cisplatino/administración & dosificación , Cisplatino/farmacocinética , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Compuestos Organoplatinos/administración & dosificación , Compuestos de Platino/farmacocinética , Ensayos Clínicos Controlados Aleatorios como Asunto
17.
Int J Nanomedicine ; 8: 2465-72, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23885173

RESUMEN

Traditional post-surgical chemotherapy for pancreatic cancer is notorious for its devastating side effects due to the high dosage required. On the other hand, legitimate concerns have been raised about nanoparticle-mediated drug delivery because of its potential cytotoxicity. Therefore, we explored the local delivery of a reduced dosage of FOLFIRINOX, a four-drug regimen comprising oxaliplatin, leucovorin, irinotecan, and fluorouracil, for pancreatic cancer using a biocompatible drug-eluting scaffold as a novel chemotherapy strategy after palliative surgery. In vitro assays showed that FOLFIRINOX in the scaffold caused massive apoptosis and thereby a decrease in the viability of pancreatic cancer cells, confirming the chemotherapeutic capability of the drug-eluting scaffold. In vivo studies in an orthotopic murine xenograft model demonstrated that the FOLFIRINOX in the scaffold had antitumorigenic and antimetastatic effects comparable with those achieved by intraperitoneal injection, despite the dose released by the scaffold being roughly two thirds lower. A mechanistic study attributed our results to the excellent ability of the FOLFIRINOX in the scaffold to destroy the CD133(+)CXCR4(+) cell population responsible for pancreatic tumorigenesis and metastasis. This clinically oriented study gives rise to a promising alternative strategy for postsurgical management of pancreatic cancer, featuring a local chemotherapeutic effect with considerable attenuation of side effects.


Asunto(s)
Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Sistemas de Liberación de Medicamentos/instrumentación , Neoplasias Pancreáticas , Análisis de Varianza , Antineoplásicos/química , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Camptotecina/análogos & derivados , Camptotecina/química , Camptotecina/farmacocinética , Camptotecina/farmacología , Supervivencia Celular/efectos de los fármacos , Citometría de Flujo , Fluorouracilo/química , Fluorouracilo/farmacocinética , Fluorouracilo/farmacología , Humanos , Irinotecán , Leucovorina/química , Leucovorina/farmacocinética , Leucovorina/farmacología , Nanofibras/química , Nanotecnología , Compuestos Organoplatinos/química , Compuestos Organoplatinos/farmacocinética , Compuestos Organoplatinos/farmacología , Oxaliplatino , Células Tumorales Cultivadas
18.
Cancer Biomark ; 13(1): 1-10, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23736016

RESUMEN

BACKGROUND: The effect of the protein S100P on biological characteristics of cancer is not clear, especially in gastric cancer. We previously showed that S100P positive gastric cancer patients have a better cumulative survival than S100P negative patients. OBJECTIVE: To study the possible mechanisms of S100P enhanced the chemosensitivity to oxaliplatin in gastric cancer cell lines. METHODS: S100P was overexpressed in vitro by plasmid transfection and downregulated by siRNA transfection in the BGC823 and SGC7901 gastric cancer cell lines. Cell survival rate, changes in the chemoresistance gene, such as GST-π, MDR1, MRP1, Topo-II, MVP and BCRP, intake of anticancer drug were measured after oxaliplatin treatment. RESULTS: In SGC7901 cells, MTT assay indicated that increased S100P expression levels decreased the survival rate and decreased S100P expression levels increased the survival rate. In BGC823 and SGC7901 cell lines, mRNA of MDR1, a chemoresistance genes, was decreased in cells that overexpressed S100P, and increased in cells with downregulation of S100P. Intracellular accumulation of platinum increased in cells with overexpressed S100P, and decreased in cells with S100P downregulation. CONCLUSIONS: S100P contributes to oxaliplatin chemosensitivity in gastric cell lines by increasing drug inflow. It might also be a novel independent prognostic factor in gastric cancer patients who receive adjuvant chemotherapy with oxaliplatin.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Unión al Calcio/biosíntesis , Proteínas de Neoplasias/biosíntesis , Compuestos Organoplatinos/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Antineoplásicos/farmacocinética , Proteínas de Unión al Calcio/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos , Humanos , Inmunohistoquímica , Proteínas de Neoplasias/genética , Compuestos Organoplatinos/farmacocinética , Oxaliplatino , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Neoplasias Gástricas/genética , Transfección
19.
Clin Exp Pharmacol Physiol ; 40(6): 371-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23556474

RESUMEN

The purpose of the present study was to determine whether copper histidine could inhibit copper transporter 1 (Ctr1)-mediated transport of oxaliplatin in vitro and thereby limit the accumulation of platinum and neurotoxicity of oxaliplatin in dorsal root ganglion (DRG) tissue in vivo. In HEK293 cells overexpressing rat Ctr1, copper histidine was shown to be transported by Ctr1 and to inhibit their Ctr1-mediated uptake of oxaliplatin. Pilot in vivo dose-finding studies showed that copper histidine at doses up to 2 mg/kg, p.o., daily for 5 days/week could be added to maximum tolerated doses of oxaliplatin (1.85 mg/kg, i.p., twice weekly) for 8 week combination treatment studies in female Wistar rats. After treatment, rats showed significant changes in sensory neuron size profiles in DRG tissue induced by oxaliplatin that were not altered by its coadministration with copper histidine. The expression of copper transporters (Ctr1 and copper-transporting P-type ATPase 1 (Atp7a)) in DRG tissue appeared unchanged following treatment with oxaliplatin given alone or with copper histidine. Platinum and copper tissue levels were higher in DRG than in most other tissues, but were unaltered by the addition of copper histidine to oxaliplatin treatment. In conclusion, copper histidine inhibited cellular uptake of oxaliplatin mediated by Ctr1 in vitro without altering the accumulation of platinum or neurotoxicity of oxaliplatin in DRG tissue in vivo at doses tolerated in combination with oxaliplatin treatment.


Asunto(s)
Proteínas de Transporte de Catión/antagonistas & inhibidores , Ganglios Espinales/efectos de los fármacos , Intoxicación del Sistema Nervioso por Metales Pesados/prevención & control , Histidina/análogos & derivados , Compuestos Organometálicos/farmacología , Compuestos Organoplatinos/toxicidad , Platino (Metal)/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Transportador de Cobre 1 , ATPasas Transportadoras de Cobre , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Femenino , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Células HEK293 , Intoxicación del Sistema Nervioso por Metales Pesados/patología , Histidina/farmacología , Humanos , Compuestos Organoplatinos/farmacocinética , Oxaliplatino , Ratas , Transfección
20.
Jpn J Radiol ; 31(6): 424-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23525952

RESUMEN

PURPOSE: To evaluate differences in the viscosity of a platinum iodized-oil suspension on the kind of platinum agent and temperature. MATERIALS AND METHODS: Viscosities of a 70 mg miriplatin and 3.5 ml iodized-oil suspension (MO suspension) and that of 100 mg cisplatin and 10 ml iodized-oil suspension (CO suspension) were evaluated at three temperatures: 25, 37, and 50 °C. Iodized-oil was used as the control. Each liquid was injected into a capillary tube and allowed to drip separately. The liquid transit time was measured, and the viscosity of each liquid was calculated at each temperature. RESULTS: The viscosity of each liquid decreased as the temperature increased: 43.3 ± 0.5, 39.2 ± 0.7, and 34.7 ± 0.6 mPa s for MO suspension, 41.3 ± 0.2, 36.9 ± 0.3, and 32.7 ± 0.9 mPa s for CO suspension, and 40.5 ± 0.2, 36.8 ± 0.2, and 33.8 ± 0.7 mPa s for iodized-oil at 25, 37, and 50 °C, respectively. The MO suspension group viscosity was significantly higher than that of the CO suspension group (p < 0.05) and the control (p < 0.05). Significant differences were found in viscosities among groups divided by temperature (25 °C-group vs. 37 °C-group, p < 0.05; 37 °C-group vs. 50 °C-group, p < 0.05). CONCLUSION: The viscosity of the platinum iodized-oil suspension can be adjusted by changing temperature.


Asunto(s)
Antineoplásicos/farmacocinética , Cisplatino/farmacocinética , Compuestos Organoplatinos/farmacocinética , Temperatura , Viscosidad , Antineoplásicos/química , Quimioembolización Terapéutica/métodos , Cisplatino/química , Emulsiones Grasas Intravenosas/farmacocinética , Humanos , Aceite Yodado/farmacocinética , Neoplasias Hepáticas/terapia , Compuestos Organoplatinos/química , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA