Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Intervalo de año de publicación
1.
Farm Hosp ; 37(2): 111-27, 2013.
Artículo en Español | MEDLINE | ID: mdl-23789755

RESUMEN

OBJECTIVE: To evaluate the influence of genetic polymorphism in UGT1A1, UGT1A7 and UGT1A9 on the population pharmacokinetics of irinotecan and its metabolites, SN-38 and SN-38G. METHODS: Plasma concentrations of irinotecan, SN-38 and SN- 38G from 72 patients were pooled to develop a population pharmacokinetic model using NONMEM VII. M3 method was used to account for plasma concentrations below the limit quantification. The effect of age, sex, body surface area, total bilirubin, co-medication, tumor type, and UGT1A1, UGT1A7 and UGT1A9 genotypes on the model parameters was evaluated. The model was internally validated using normalized visual predictive check (NVPC) and normalized predictive distribution errors (NPDE). RESULTS: The typical values (between-subject variability; %) of the irinotecan, SN-38 and SN-38G clearances were 42,9 L/h (56,4%), 1340 L/h (76,8%) and 188 L/h (70,1%), respectively. The presence of UGT1A1*28, UGT1A7*3, UGT1A9*22 genotypes decreases SN-38 clearance between 20 and 36%. Internal validation confirms the population pharmacokinetic model describe the time course of irinotecan, SN-38 and SN-38G plasma concentration and their associated variability in cancer patients. CONCLUSION: The inclusion of pharmacokinetic-pharmacogenomic information can add value to the individualized dose adjustment of irinotecan, because it will let quantitatively handle dose reductions in patients with iatrogenic toxicity due to UGT1A1 genetic polymorphisms.


Objetivo: Evaluar la influencia de los polimorfismos genéticos en UGT1A1, UGT1A7 y UGT1A9 sobre la farmacocinética poblacional de irinotecán y sus metabolitos, SN-38 y SN-38G. Metodología: Las concentraciones plasmáticas de irinotecán, SN-38 y SN-38G determinadas en 72 pacientes se utilizaron para desarrollar un modelo farmacocinético poblacional en el programa NONMEM VII. Se empleó el método M3 para incluir en el análisis las concentraciones por debajo del límite de cuantificación de la técnica analítica. Se evaluó el efecto de la edad, sexo, superficie corporal, bilirrubina total, medicación concomitante, tipo de tumor y polimorfismos genéticos en UGT1A1, UGT1A7 y UGT1A9 sobre los parámetros farmacocineticos del modelo. La validación interna del modelo farmacocinético se realizó mediante normalized visual predictive check (NVPC) y normalized predictive distribution error (NPDE). Resultados: El valor medio (variabilidad interpaciente, %) del aclaramiento de irinotecán, SN-38 y SN-38G ha sido 42,9 (56,4%), 1340 (76,8%) y 188 L/h (70,1%), respectivamente. La presencia de alelos con baja actividad enzimática (UGT1A1*28, UGT1A7*3 y UGT1A9*22) redujo el aclaramiento de SN-38 entre un 20 y un 36%. La validación interna ha confirmado que el modelo farmacocinético poblacional resulta adecuado para describir la evolución temporal de las concentraciones plasmáticas de irinotecán, SN-38 y SN-38G y su variabilidad en pacientes oncológicos. Conclusión: La inclusión de información farmacocinética-farmacogenética puede añadir valor a la personalización de la dosificación de irinotecán por cuanto que permitirá manejar cuantitativamente las reducciones de dosis en pacientes con toxicidad iatrogénica debido a los polimorfismos genéticos en UGT1A1.


Asunto(s)
Antineoplásicos Fitogénicos/farmacocinética , Camptotecina/análogos & derivados , Glucuronatos/farmacocinética , Glucuronosiltransferasa/genética , Polimorfismo Genético , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos Fitogénicos/sangre , Antineoplásicos Fitogénicos/uso terapéutico , Camptotecina/sangre , Camptotecina/farmacocinética , Camptotecina/uso terapéutico , Femenino , Glucuronatos/sangre , Glucuronatos/uso terapéutico , Humanos , Irinotecán , Masculino , Persona de Mediana Edad , Neoplasias/tratamiento farmacológico , Neoplasias/genética , UDP Glucuronosiltransferasa 1A9
2.
Farm. hosp ; 37(2): 111-127, mar.-abr. 2013. ilus, tab
Artículo en Español | IBECS | ID: ibc-115662

RESUMEN

Objetivo: Evaluar la influencia de los polimorfismos genéticos en UGT1A1, UGT1A7 y UGT1A9 sobre la farmacocinética poblacional de irinotecán y sus metabolitos, SN-38 y SN-38G. Metodología: Las concentraciones plasmáticas de irinotecán, SN-38 y SN-38G determinadas en 72 pacientes se utilizaron para desarrollar un modelo farmacocinético poblacional en el programa NONMEM VII. Se empleó el método M3 para incluir en el análisis las concentraciones por debajo del límite de cuantificación de la técnica analítica. Se evaluó el efecto de la edad, sexo, superficie corporal, bilirrubina total, medicación concomitante, tipo de tumor y polimorfismos genéticos en UGT1A1, UGT1A7 y UGT1A9 sobre los parámetros farmacocineticos del modelo. La validación interna del modelo farmacocinético se realizó mediante normalized visual predictive check(NVPC) y normalized predictive distribution error (NPDE). Resultados: El valor medio (variabilidad interpaciente, %) del aclaramiento de irinotecán, SN-38 y SN-38G ha sido 42,9 (56,4%), 1340 (76,8%) y 188 L/h (70,1%), respectivamente. La presencia de alelos con baja actividad enzimática (UGT1A1*28, UGT1A7*3 y UGT1A9*22) redujo el aclaramiento de SN-38 entre un 20 y un 36%. La validación interna ha confirmado que el modelo farmacocinético poblacional resulta adecuado para describir la evolución temporal de las concentraciones plasmáticas de irinotecán, SN-38 y SN-38G y su variabilidad en pacientes oncológicos. Conclusión: La inclusión de información farmacocinética-farmacogenética puede añadir valor a la personalización de la dosificación de irinotecán por cuanto que permitirá manejar cuantitativamente las reducciones de dosis en pacientes con toxicidad iatrogénica debido a los polimorfismos genéticos en UGT1A1 (AU)


Objective: To evaluate the Influence of genetic polymorphism in UGT1A1, UGT1A7 and UGT1A9 on the population pharmacokinetics of irinotecan and its metabolites, SN-38 and SN-38G. Methods: Plasma concentrations of irinotecan, SN-38 and SN-38G from 72 patients were pooled to develop a population pharmacokinetic model using NONMEM VII. M3 method was used to account for plasma concentrations below the limit quantification. The effect of age, sex, body surface area, total bilirubin, comedication, tumor type, and UGT1A1, UGT1A7 and UGT1A9 genotypes on the model parameters was evaluated. The model was internally validated using normalized visual predictive check (NVPC) and normalized predictive distribution errors (NPDE). Results: The typical values (between-subject variability; %) of the irinotecan, SN-38 and SN-38G clearances were 42,9 L/h (56,4%), 1340 L/h (76,8%) and 188 L/h (70,1%), respectively. The presence of UGT1A1*28, UGT1A7*3, UGT1A9*22 genotypes decreases SN-38 clearance between 20 and 36%. Internal validation confirms the population pharmacokinetic model describe the time course of irinotecan, SN-38 and SN-38G plasma concentration and their associated variability in cancer patients. Conclusion: The inclusion of pharmacokinetic-pharmacogenomic information can add value to the individualized dose adjustment of irinotecan, because it will let quantitatively handle dose reductions in patients with iatrogenic toxicity due to UGTIAVs genetic polymorphisms (AU)


Asunto(s)
Humanos , Inhibidores de Topoisomerasa I/farmacocinética , /genética , Marcadores Genéticos , Polimorfismo Genético , Técnicas de Genotipaje/métodos
3.
Clin Transl Oncol ; 13(11): 812-8, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22082647

RESUMEN

INTRODUCTION Epidermal growth factor receptor (EGFR) mutation related to tyrosine kinase inhibitors' (TKIs) responsiveness in non-small cell lung cancer (NSCLC) has become an important issue for therapeutic decision-making in NSCLC patients. MATERIAL AND METHODS Sixty-nine Caucasian NSCLC patients were screened for mutations in the tyrosine kinase (TK) domain of EGFR by direct sequencing from December 2005 to September 2010. RESULTS Activating mutations in the EGFR TK domain were found in 8 of 69 (11.6%) (7 deletions in exon 19 and one L858R mutation in exon 21). Seven of those mutations were found in adenocarcinoma and one mutation in bronchiolo-alveolar carcinoma; five of them in females (one smoker) and three of them in males (one smoker). All patients carrying activating mutations in the TK domain of EGFR were treated with TKIs. Ten patients not carrying an activating mutation in EGFR, who progressed after chemotherapy, were also treated with compassionate use of EGFR-specific TKIs (gefitinib or erlotinib). An objective response (partial response) was observed in all patients carrying an activating mutation in EGFR that received TKIs. Median overall survival for these patients has not been reached, however mean survival has been estimated at 39.5 months (95% CI, 22-57). CONCLUSIONS As previously reported, EGFR TK mutational analysis was a predictive test for response to targeted therapy with EGFR TKIs. The early identification of these patients consistently attains disease response and clearly improves outcomes.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/genética , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Análisis Mutacional de ADN , Resistencia a Antineoplásicos/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib , Femenino , Gefitinib , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/enzimología , Masculino , Persona de Mediana Edad , Quinazolinas/uso terapéutico , Análisis de Supervivencia
4.
Clin. transl. oncol. (Print) ; 13(11): 812-818, nov. 2011. tab, ilus
Artículo en Inglés | IBECS | ID: ibc-125943

RESUMEN

INTRODUCTION Epidermal growth factor receptor (EGFR) mutation related to tyrosine kinase inhibitors' (TKIs) responsiveness in non-small cell lung cancer (NSCLC) has become an important issue for therapeutic decision-making in NSCLC patients. MATERIAL AND METHODS Sixty-nine Caucasian NSCLC patients were screened for mutations in the tyrosine kinase (TK) domain of EGFR by direct sequencing from December 2005 to September 2010. RESULTS Activating mutations in the EGFR TK domain were found in 8 of 69 (11.6%) (7 deletions in exon 19 and one L858R mutation in exon 21). Seven of those mutations were found in adenocarcinoma and one mutation in bronchiolo-alveolar carcinoma; five of them in females (one smoker) and three of them in males (one smoker). All patients carrying activating mutations in the TK domain of EGFR were treated with TKIs. Ten patients not carrying an activating mutation in EGFR, who progressed after chemotherapy, were also treated with compassionate use of EGFR-specific TKIs (gefitinib or erlotinib). An objective response (partial response) was observed in all patients carrying an activating mutation in EGFR that received TKIs. Median overall survival for these patients has not been reached, however mean survival has been estimated at 39.5 months (95% CI, 22-57). CONCLUSIONS As previously reported, EGFR TK mutational analysis was a predictive test for response to targeted therapy with EGFR TKIs. The early identification of these patients consistently attains disease response and clearly improves outcomes (AU)


Asunto(s)
Humanos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Resistencia a Antineoplásicos/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/enzimología , Mutación , Quinazolinas/uso terapéutico , Análisis de Supervivencia
5.
Clin Transl Oncol ; 10(7): 442-4, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18628075

RESUMEN

Most of the somatic epidermal growth factor receptor (EGFR) mutations described to date in non-smallcell lung cancer (NSCLC) patients are located in the kinase domain and are considered activating mutations. Some of these mutations are associated with response to specific EGFR tyrosine kinase inhibitors (TKI) such as gefitinib and erlotinib. Here we report a case of a previously undescribed EGFR nonsense mutation in a lung adenocarcinoma patient who did not derive any clinical benefit with combination chemotherapy and erlotinib. To the best of our knowledge this is the second report in the literature describing an EGFR nonsense mutation in lung cancer patients.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinazolinas/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Secuencia de Bases , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Codón sin Sentido , Clorhidrato de Erlotinib , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Neoplasias Pulmonares/tratamiento farmacológico , Masculino , Persona de Mediana Edad
6.
Clin. transl. oncol. (Print) ; 10(7): 442-444, jul. 2008. ilus
Artículo en Inglés | IBECS | ID: ibc-123477

RESUMEN

Most of the somatic epidermal growth factor receptor (EGFR) mutations described to date in non-smallcell lung cancer (NSCLC) patients are located in the kinase domain and are considered activating mutations. Some of these mutations are associated with response to specific EGFR tyrosine kinase inhibitors (TKI) such as gefitinib and erlotinib. Here we report a case of a previously undescribed EGFR nonsense mutation in a lung adenocarcinoma patient who did not derive any clinical benefit with combination chemotherapy and erlotinib. To the best of our knowledge this is the second report in the literature describing an EGFR nonsense mutation in lung cancer patients (AU)


No disponible


Asunto(s)
Humanos , Femenino , Persona de Mediana Edad , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Quinazolinas/uso terapéutico , Receptores ErbB/genética , /uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Secuencia de Bases , Codón sin Sentido , Inmunohistoquímica/métodos , Inmunohistoquímica , Hibridación Fluorescente in Situ
8.
Ciba Found Symp ; 212: 24-35; discussion 35-41, 42-5, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9524762

RESUMEN

HGF/SF and HGF1/MSP define a novel growth factor family whose members share the domain structure and the proteolytic process of activation of the blood proteinase precursor plasminogen. The amino acid and RNA sequences of HGF/SF and HGF1/MSP, the intron-exon organization of their genes and the predicted 3D structure of individual domains indicate that HGF/SF and HGF1/MSP evolved along with plasminogen and other members of the kringle-serine proteinase (KSP) superfamily from an ancestral gene that contained a single copy of the kringle domain, a serine proteinase domain and an activation peptide connecting the two domains. A series of intragenic duplications of the kringle domain, gene duplications, exon shuffling and deletions is responsible for the genes currently present in mammals, avians and amphibians. Plasminogen, HGF/SF and HGF1/MSP represent paradigmatic examples of the modern, multi-domain proteins typically associated with vertebrate organisms and illustrate a novel evolutionary pathway that led to the emergence of molecules with growth regulatory activity from proteolytic enzymes.


Asunto(s)
Evolución Biológica , Sustancias de Crecimiento/química , Factor de Crecimiento de Hepatocito/química , Plasminógeno/química , Precursores de Proteínas/química , Proteínas Proto-Oncogénicas , Secuencia de Aminoácidos , Animales , Humanos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
9.
Eur J Cancer ; 32A(12): 2136-41, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9014757

RESUMEN

Prior studies have shown that, in some human tumour cells, increased expression of the multidrug resistance gene MDR1 can be induced in response to certain stress conditions such as a transient exposure to cytotoxic agents. Little is known about the possibility of increasing the expression of the recently cloned multidrug resistance-associated protein (MRP) in response to a transient exposure to cytotoxic drugs. In order to examine this possibility, we have used sensitive assays (RT-PCR, flow cytometry) and the sensitive large cell lung cancer cell line, COR-L23/P, and the revertant line (COR-L23/Rev), generated by growing the doxorubicin-selected, MRP-overexpressing resistant variant COR-L23/R without drug exposure for 24-28 weeks. COR-L23/Rev overexpresses MRP, but to a lesser extent than COR-L23/R. COR-L23/Rev rapidly recovered similar levels of MRP mRNA, protein expression, resistance and drug accumulation deficit as COR-L23/R after a 48-72 h exposure to cytotoxic concentrations of doxorubicin or vincristine but not cisplatin. The increase in MRP mRNA could only be detected 3 to 4 days after the transient exposure to drugs. However, when the parental line, COR-L23/P, was exposed to equitoxic doses of doxorubicin, vincristine or cisplatin, no increase in the levels of MRP mRNA could be observed at higher doses (5- to 10-fold the IC50) of doxorubicin or vincristine (but not of cisplatin), we detected a transient increase in the levels of MDR1 mRNA immediately after short-term exposure. In conclusion, we have shown that a human revertant lung cancer cell line (COR-L23/Rev) has the ability to recover quickly, similar levels of MRP expression and resistance as COR-L23/R after a transient exposure to the MDR-drugs doxorubicin and vincristine.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes MDR , Neoplasias Pulmonares/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Cisplatino/farmacología , Doxorrubicina/farmacología , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Humanos , Neoplasias Pulmonares/metabolismo , Fenotipo , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Neoplásico/genética , Células Tumorales Cultivadas/efectos de los fármacos , Vincristina/farmacología
10.
Br J Cancer ; 72(5): 1294-9, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7577485

RESUMEN

In order to test whether circumvention of clinical resistance can be obtained in common solid tumours by targeting different drug resistance mechanisms, a phase I clinical and immunological study was designed. The purpose of the study was to determine the dose of cyclosporin A (CsA), in combination with doxorubicin (DOX) and ifosfamide (IFX), needed to achieve steady-state whole-blood levels of 2000 ng ml-1 and the associated toxicity of this combination. Treatment consisted of CsA 5 mg kg-1 as a 2 h loading infusion, followed by a CsA 3 day continuous infusion (c.i.) (days 1-3) at doses that were escalated from 10 to 18 mg kg-1 day-1. Chemotherapy consisted of DOX 55 mg m-2 by i.v. 24 h c.i. (day 2) and IFX 2 g m-2 i.v. over 1 h on days 1 and 3. Treatments were repeated every 4 weeks. Eighteen patients with previously treated resistant solid tumours received 39 cycles. Mean steady-state CsA levels > or = 2000 ng ml-1 were reached at 5 mg kg-1 loading dose followed by a 3 day c.i. of 16 mg kg-1 day-1 or greater. Haematological toxicity was greater than expected for the same chemotherapy alone. One patient died of intracranial haemorrhage due to severe thrombopenia. Other observed toxicities were: asymptomatic hyperbilirubinaemia (46% cycles), mild nephrotoxicity (20% cycles), hypomagnesaemia (72% cycles), mild increase in body weight (100% cycles), hypertension (15% cycles) and headache (15% cycles). Overall the toxicity was acceptable and manageable. No alterations in absolute lymphocyte number, the lymphocyte subsets studied (CD3, CD4, CD8, CD19) or CD4/CD8 ratio were observed in patients receiving more than one treatment cycle, although there were significant and non-uniform variations in the values of the different lymphocyte subsets studied when pre- and post-treatment values were compared. There was also a significant increase in the CD4/CD8 ratio. Tumour regressions were observed in two patients (epidermoid carcinoma of the cervix and Ewing's sarcoma). The CsA dose recommended for phase II trials is a 5 mg kg-1 loading dose followed by a 3-day c.i. of 16 mg kg-1 day-1 simultaneously with DOX and IFX at the doses administered in this study.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ciclosporina/uso terapéutico , Inmunidad Celular/efectos de los fármacos , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Unión Competitiva , Ciclosporina/efectos adversos , Ciclosporina/farmacocinética , Ciclosporina/farmacología , Doxorrubicina/administración & dosificación , Doxorrubicina/efectos adversos , Doxorrubicina/farmacocinética , Resistencia a Antineoplásicos , Femenino , Humanos , Ifosfamida/administración & dosificación , Ifosfamida/efectos adversos , Ifosfamida/farmacocinética , Recuento de Linfocitos/efectos de los fármacos , Subgrupos Linfocitarios/efectos de los fármacos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteínas de Neoplasias/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Inducción de Remisión
12.
Am J Clin Oncol ; 16(4): 332-7, 1993 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8328412

RESUMEN

Amphotericin B enhances the cytotoxicity of certain antineoplastic agents in vitro and in vivo. A phase II study was designed to evaluate whether this effect can be produced in the treatment of metastatic soft-tissue sarcomas (STS). The program AIDAB consisted of ADM 50 mg/m2 i.v. bolus at day 1; ifosfamide (IFX) 1.5 g/m2 in 1 hour i.v. infusion/day x 3 days; mesna 300 mg/m2 i.v. bolus before and 4 and 8 hours after IFX; dacarbazine (DTIC) 400 mg/m2 i.v. in 6 hours infusion/day x 3 days; and amphotericin B 25 mg/m2 i.v. in 6 hours infusion/day x 3 days; repeated every 4 weeks. There were 25 patients evaluable for response and toxicity, 22 with no previous chemotherapy. The median age was 44, (range: 16-64); 14 males, 11 females with a Karnofsky range of 40 to 90%. The response rate was 48% (4% complete response and 44% partial response). The median duration of response was 6.6 months. Of these 25 patients, 17 (68%) have died; the median survival was 12 months (range: 3-51 months). A total of 175 cycles were given to the 25 patients, with a mean of 7 cycles per patient. Toxicities encountered were leukopenia and/or thrombocytopenia, grade IV (WHO), in 9 patients (36%); fever and chills during amphotericin B infusion in 40%; vomiting, grade III (WHO), in 56%; mild mucositis in 12%, and a symptomatic decrease in cardiac ejection fraction in one patient. There was one toxic death due to severe thrombocytopenia. We can conclude that AIDAB is effective in the treatment of metastatic soft-tissue sarcoma. The addition of amphotericin B did not improve the response rate or survival above what is expected from chemotherapy alone.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Sarcoma/tratamiento farmacológico , Sarcoma/secundario , Neoplasias de los Tejidos Blandos/tratamiento farmacológico , Neoplasias de los Tejidos Blandos/secundario , Adolescente , Adulto , Anfotericina B/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Dacarbazina/administración & dosificación , Doxorrubicina/administración & dosificación , Femenino , Humanos , Ifosfamida/administración & dosificación , Masculino , Persona de Mediana Edad , Proyectos Piloto , Inducción de Remisión , Análisis de Supervivencia
13.
Rev Med Univ Navarra ; 38(1): 13-8, 1993.
Artículo en Español | MEDLINE | ID: mdl-8711305

RESUMEN

Thirty previously untreated patients with advanced germ cell testicular tumours received PVB or BEP. Four patients with bulky disease underwent debulking surgery before initiating chemotherapy and two between cycles of treatment. Twenty-seven (90%) complete responses, two (6.7%) partial responses and one (3.3%) no changes were observed. Both partial remissions were rendered disease-free with surgical removal of residual disease. Four patients presented tumour progression with PVB or BEP, three of whom developed a non-germ cell malignancy within the germ cell tumour. One toxic death and three patients with radiological evidence of reversible interstitial pneumonitis attributed to bleomycin were observed. With a median follow-up time of 38.5 months, range 2.5 to 130 months, 83.3% of the patients are alive and free of disease. Actuarial overall survival is 85.6%. This study confirms once again the high percentage of curability of this disease.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Seminoma/tratamiento farmacológico , Neoplasias Testiculares/tratamiento farmacológico , Adolescente , Adulto , Anciano , Bleomicina/administración & dosificación , Bleomicina/efectos adversos , Cisplatino/administración & dosificación , Cisplatino/efectos adversos , Terapia Combinada , Etopósido/administración & dosificación , Etopósido/efectos adversos , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Orquiectomía , Seminoma/patología , Seminoma/cirugía , Análisis de Supervivencia , Neoplasias Testiculares/patología , Neoplasias Testiculares/cirugía , Vinblastina/administración & dosificación , Vinblastina/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA