Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Am Pharm Assoc (2003) ; 62(4): 1430-1437, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35461778

RESUMEN

BACKGROUND: Previous large-scale vaccination clinics have been successful before the coronavirus disease 2019 (COVID-19) pandemic; however, owing to the strict storage requirements and pharmaceutical preparation needed for the COVID-19 vaccines, careful thought and planning were necessary to successfully deploy these clinics immediately after vaccine availability. The focus of this manuscript is to describe the development and implementation of COVID-19 vaccination clinics in a large public university, using professionals from within and outside of its health sciences schools. OBJECTIVES: The primary objective of this project was to (1) implement COVID-19 vaccination clinics for university faculty, staff, students, and community members. Additional objectives of the clinics were to (2) actively incorporate pharmacy, nursing, and medical students into the clinic workflow; (3) promote interprofessional collaboration among faculty and students; and (4) assess patient satisfaction. PRACTICE DESCRIPTION: The School of Pharmacy faculty, in conjunction with the Office of Strategic Initiatives, planned and coordinated COVID-19 vaccination clinics from December 2020 to July 2021. Students and faculty from schools of pharmacy, nursing, and medicine were used. COVID-19 vaccinations were offered to university faculty, staff, and students and community members based on the Centers for Disease Control and Prevention priority groups. The clinic processes were designed such that they could be scaled from 100 to 2,000 participants per day. PRACTICE INNOVATION: The School of Pharmacy led approach was adjustable depending on the number of patients, continuously monitored and adaptable. The importance of pharmacists as part of the interprofessional health care team was exemplified by faculty and students involved. EVALUATION METHODS: All patients receiving COVID-19 vaccinations at the clinics were e-mailed anonymous surveys for assessment of the quality of the vaccination encounter after completion of their primary vaccine series. RESULTS: More than 15,000 COVID-19 vaccinations were provided through the clinics from December 2020 to July 2021. Professional staffing totaled 3352 hours for the 48 clinics. Thirty-eight percent of the vaccinated patients responded to the clinic satisfaction survey with predominately excellent ratings. CONCLUSION: COVID-19 vaccination clinics can be successfully planned and implemented in a scalable fashion in a large university setting using an interprofessional team approach.


Asunto(s)
COVID-19 , Servicios Farmacéuticos , COVID-19/prevención & control , Vacunas contra la COVID-19 , Humanos , Farmacéuticos , Universidades , Vacunación
2.
Cancer Invest ; 29(9): 617-25, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22011284

RESUMEN

We conducted a phase I study to assess safety, pharmacokinetics, pharmacodynamics, and activity of lonafarnib plus gemcitabine. Subjects received oral lonafarnib twice daily and gemcitabine on days 1, 8, and 15 every 28 days; multiple dose levels were explored. Lonafarnib had no apparent effect on gemcitabine PK. Mean lonafarnib half-life ranged from 4 to 7 hr; median T(max) values ranged from 4 to 8 hr. Two patients had partial response; seven patients had stable disease at least 6 months. Oral lonafarnib at 150 mg a.m./100 mg p.m. plus gemcitabine at 1,000 mg/m(2) is the maximum tolerated dose with acceptable safety and tolerability.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias/tratamiento farmacológico , Administración Oral , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Desoxicitidina/administración & dosificación , Desoxicitidina/efectos adversos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacocinética , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Neoplasias/metabolismo , Piperidinas/administración & dosificación , Piperidinas/efectos adversos , Piperidinas/farmacocinética , Piridinas/administración & dosificación , Piridinas/efectos adversos , Piridinas/farmacocinética , Gemcitabina
3.
Int J Radiat Oncol Biol Phys ; 70(4): 1066-72, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17881149

RESUMEN

PURPOSE: Concurrent chemotherapy and radiation therapy (RT) are used to treat patients with esophageal cancer. The optimal combination of chemotherapeutic agents with RT is not well established. We evaluated the safety and preliminary efficacy of a combination of UFT/leucovorin, carboplatin, and paclitaxel with RT in a Phase I study of patients with advanced esophageal cancer. METHODS AND MATERIALS: Patients with squamous cell carcinoma or adenocarcinoma of the esophagus initially received UFT/leucovorin, carboplatin, and paclitaxel with RT (1.8 Gy daily to 45 Gy). After completion, the disease was restaged and patients were evaluated for surgery. Primary end points included determination of dose-limiting toxicities (DLTs) and a recommended Phase II dose. Secondary objectives included determination of non-DLTs, as well as preliminary radiographic and pathologic response rates. RESULTS: Twelve patients were enrolled (11 men, 1 woman). All were assessable for toxicity and efficacy. One of 6 patients at Dose Level 1 (UFT/leucovorin, 200/30 mg twice daily on RT days; carboplatin, area under the curve [AUC] 5, Weeks 1 and 4; paclitaxel, 175 mg/m2 Weeks 1 and 4) had a DLT (febrile neutropenia). Of these 6 patients, 4 underwent esophagectomy and none achieved a pathologic complete response. Six patients were then enrolled at Dose Level 2 (UFT/leucovorin, 300/30 mg in the morning and 200/30 mg in the evening on RT days; carboplatin, AUC 5, Weeks 1 and 4; paclitaxel, 175 mg/m2 Weeks 1 and 4). Two of 6 patients at Dose Level 2 developed DLTs (febrile neutropenia in both). Esophagectomy was performed in 3 patients, with 2 achieving a pathologic complete response. CONCLUSIONS: Maximum tolerated doses in this study were UFT/leucovorin, 200/30 mg twice daily on RT days; carboplatin, AUC 5, Weeks 1 and 4; and paclitaxel, 175 mg/m2 Weeks 1 and 4 when delivered with external RT. In this small study, this regimen appears active, but toxic.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/radioterapia , Unión Esofagogástrica , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Adenocarcinoma/radioterapia , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carboplatino/administración & dosificación , Carboplatino/efectos adversos , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/radioterapia , Combinación de Medicamentos , Neoplasias Esofágicas/patología , Femenino , Humanos , Leucovorina/administración & dosificación , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Paclitaxel/administración & dosificación , Paclitaxel/efectos adversos , Dosificación Radioterapéutica , Tegafur/administración & dosificación , Tegafur/efectos adversos , Uracilo/administración & dosificación , Uracilo/efectos adversos
4.
J Pharmacol Exp Ther ; 327(3): 770-6, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18791061

RESUMEN

Elimination of hydrogen sulfide from glutathione (GSH) converts a well known cellular nucleophile to an electrophilic species, gamma-glutamyldehydroalanylglycine (EdAG). We have found that a sulfonium metabolite formed from GSH and busulfan undergoes a facile beta-elimination reaction to give EdAG, which is an alpha,beta-unsaturated dehydroalanyl analog of GSH. EdAG was identified as a metabolite of busulfan in a human liver cytosol fraction. EdAG condenses with GSH in a Michael addition reaction to produce a lanthionine thioether [(2-amino-5-[[3-[2-[[4-amino-5-hydroxy-5-oxopentanoyl]amino]-3-(carboxymethylamino)-3-oxopropyl]sulfanyl-1-(carboxymethylamino)-1-oxopropan-2-yl]amino]-5-oxopentanoic acid); GSG], which is a nonreducible analog of glutathione disulfide. EdAG was less cytotoxic than busulfan to C6 rat glioma cells. GSH and EdAG were equally effective in displacing a glutathione S-transferase (GST) isozyme (human GSTA1-1) from a GSH-agarose column. The finding of an electrophilic metabolite of GSH suggests that alteration of cellular GSH concentrations, irreversible nonreducible glutathionylation of proteins, and interference with GST function may contribute to the toxicity of busulfan.


Asunto(s)
Busulfano/metabolismo , Glutatión Transferasa/metabolismo , Glutatión/análogos & derivados , Glutatión/metabolismo , Alanina/análogos & derivados , Animales , Antineoplásicos Alquilantes , Busulfano/farmacología , Línea Celular Tumoral , Glioma/tratamiento farmacológico , Disulfuro de Glutatión/análogos & derivados , Humanos , Hígado , Ratas
5.
Drug Metab Dispos ; 36(8): 1546-52, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18474673

RESUMEN

The present work documents the first example of an enzyme-catalyzed beta-elimination of a thioether from a sulfonium cysteine S-conjugate. beta-(S-Tetrahydrothiophenium)-L-alanine (THT-A) is the cysteine S-conjugate of busulfan. THT-A slowly undergoes a nonenzymatic beta-elimination reaction at pH 7.4 and 37 degrees C to yield tetrahydrothiophene, pyruvate, and ammonia. This reaction is accelerated by 1) rat liver, kidney, and brain homogenates, 2) isolated rat liver mitochondria, and 3) pyridoxal 5'-phosphate (PLP). A PLP-dependent enzyme in rat liver cytosol that catalyzes a beta-lyase reaction with THT-A was identified as cystathionine gamma-lyase. This unusual drug metabolism pathway represents an alternate route for intermediates in the mercapturate pathway.


Asunto(s)
Antineoplásicos Alquilantes/farmacocinética , Busulfano/farmacocinética , Cisteína/metabolismo , Liasas/metabolismo , Animales , Encéfalo/enzimología , Encéfalo/metabolismo , Riñón/enzimología , Riñón/metabolismo , Hígado/enzimología , Hígado/metabolismo , Fosfato de Piridoxal/metabolismo , Ratas
6.
Cancer Chemother Pharmacol ; 59(1): 139-47, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16819636

RESUMEN

PURPOSE: To assess the maximum tolerated dose, safety, and pharmacokinetic (PK) profile of escalating doses of the novel topoisomerase I (topo I) inhibitor edotecarin (J-107088) given as a 2-h intravenous (IV) infusion once every 21 days in patients with advanced solid tumors who had not responded to standard therapy. PATIENTS AND METHODS: Twenty-nine patients (18M:11F) received a 2-h IV infusion of edotecarin in doses of 6, 8, 11, 13, or 15 mg/m(2) every 21 days (with an additional 1-2 weeks permitted for recovery) and were evaluated for safety, PK, and tumor response. RESULTS: The most common non-hematologic toxicities were grade 1-2 nausea, fatigue, anorexia, vomiting, and fever. The most common hematologic toxicities were grade 1-2 thrombocytopenia and grade 3-4 neutropenia, leukopenia, and anemia. No grade 3-4 diarrhea was reported. Dose-limiting toxicities were observed in four patients at the 15 mg/m(2) dose and one patient at the 13 mg/m(2) dose. These toxicities included grade 3 nausea, vomiting, headache, and fatigue, as well as grade 4 neutropenia and febrile neutropenia. The maximum tolerated dose was declared at 15 mg/m(2). One patient with bladder cancer had a confirmed partial response at a dose of 13 mg/m(2). There was a trend to dose-proportional increases in edotecarin peak plasma concentrations and area under the curve values. Renal excretion of edotecarin was minimal (3-8% of the dose). CONCLUSION: The recommended Phase II dose of edotecarin is 13 mg/m(2) once every 21 days. The toxicities in this study were those typical of cytotoxic chemotherapy and less severe than those associated with other topo I inhibitors. The observed safety profile and preliminary evidence of clinical benefit warrant further investigation of this drug as monotherapy or part of combination therapy in patients with solid tumors.


Asunto(s)
Carbazoles/efectos adversos , Carbazoles/farmacocinética , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/farmacocinética , Indoles/efectos adversos , Indoles/farmacocinética , Neoplasias/metabolismo , Inhibidores de Topoisomerasa I , Adulto , Anciano , Anciano de 80 o más Años , Área Bajo la Curva , Recuento de Células Sanguíneas , Carbazoles/uso terapéutico , Cromatografía Líquida de Alta Presión , Estudios de Cohortes , Relación Dosis-Respuesta a Droga , Fatiga/inducido químicamente , Fatiga/epidemiología , Femenino , Fiebre/inducido químicamente , Fiebre/epidemiología , Enfermedades Hematológicas/inducido químicamente , Enfermedades Hematológicas/epidemiología , Humanos , Indoles/uso terapéutico , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Náusea/inducido químicamente , Náusea/epidemiología , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Neutropenia/inducido químicamente , Neutropenia/epidemiología , Vómitos/inducido químicamente , Vómitos/epidemiología
7.
Clin Cancer Res ; 12(5): 1540-6, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16533779

RESUMEN

PURPOSE: A phase I trial of intrathecal Spartaject Busulfan (SuperGen, Inc., San Ramon, CA) was conducted in children with neoplastic meningitis following recurrent primary brain tumors to describe toxicities, estimate the maximum tolerated dose (MTD), and document evidence of responses to this agent. EXPERIMENTAL DESIGN: The continuous reassessment method was used to assign cohorts of patients to doses of intrathecal Spartaject Busulfan via an Ommaya reservoir and/or lumbar puncture twice weekly for 2 weeks followed by an assessment of toxicity and response. Patients with stable disease or an objective response continued to receive intrathecal Spartaject Busulfan plus systemic chemotherapy at regular intervals. Cerebrospinal fluid and blood were obtained for pharmacokinetic studies in patients with Ommaya reservoirs after the first dose of intrathecal Spartaject Busulfan. Seven evaluable patients were assigned to the starting dose of 5 mg, two patients to 7.5 mg, three patients to 10 mg, seven patients to 13 mg, and four patients to 17 mg. RESULTS: Between September 2000 and May 2003, 28 patients were enrolled in this study. Twenty-three patients (median age, 8.8 years; range, 2.5-19.5 years) were evaluable for estimating the MTD, and dose-limiting toxicities were observed in three and included grade 3 vomiting (n = 1 at 5 mg), grade 3 headache (n = 1 at 17 mg), and grade 3 arachnoiditis (n = 1 at 17 mg). Pharmacokinetic data showed that post-infusion concentrations of busulfan ranged from 50 to 150 microg/mL and declined to <1 microg/mL within 5 hours. CONCLUSIONS: Intrathecal Spartaject Busulfan was well tolerated in children with neoplastic meningitis from brain tumors, and the recommended dose for future phase II studies is 13 mg.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Busulfano/uso terapéutico , Neoplasias Meníngeas/tratamiento farmacológico , Meningitis/tratamiento farmacológico , Adolescente , Adulto , Antineoplásicos Alquilantes/farmacocinética , Busulfano/farmacocinética , Niño , Preescolar , Neoplasias del Plexo Coroideo/sangre , Neoplasias del Plexo Coroideo/líquido cefalorraquídeo , Neoplasias del Plexo Coroideo/tratamiento farmacológico , Estudios de Cohortes , Ependimoma/sangre , Ependimoma/líquido cefalorraquídeo , Ependimoma/tratamiento farmacológico , Femenino , Glioma/sangre , Glioma/líquido cefalorraquídeo , Glioma/tratamiento farmacológico , Humanos , Inyecciones Espinales , Masculino , Dosis Máxima Tolerada , Neoplasias Meníngeas/sangre , Neoplasias Meníngeas/líquido cefalorraquídeo , Meningitis/sangre , Meningitis/líquido cefalorraquídeo , Tumores Neuroectodérmicos Primitivos/sangre , Tumores Neuroectodérmicos Primitivos/líquido cefalorraquídeo , Tumores Neuroectodérmicos Primitivos/tratamiento farmacológico
8.
Clin Cancer Res ; 12(15): 4636-40, 2006 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16899612

RESUMEN

PURPOSE: The herbal supplement garlic (Allium sativum) is commonly used by cancer patients. Preclinical studies have shown that allicin, a major component of garlic, may affect cytochrome P450 3A4 (CYP3A4) activity. This study examines the influence of garlic supplementation on the pharmacokinetics of docetaxel, a CYP3A4 substrate. EXPERIMENTAL DESIGN: Women with metastatic breast cancer were treated with docetaxel (30 mg/m(2)) given weekly for 3 of 4 weeks. Three days after the initial dose of docetaxel, patients received 600 mg of garlic twice daily for 12 consecutive days. Docetaxel pharmacokinetics were assessed during the first three administrations. RESULTS: In 10 evaluable patients, the mean baseline clearance of docetaxel was 30.8 L/h/m(2) [95% confidence intervals (95% CI), 16.7-44.9]. Coadministration of garlic reduced mean clearance of docetaxel to 23.7 L/h/m(2) (95% CI, 15.5-31.8) and 20.0 L/h/m(2) (95% CI, 13.3-26.7) on days 8 and 15, respectively (P = 0.17). Additional pharmacokinetic variables of docetaxel, including peak concentration (P = 0.79), area under the curve (P = 0.36), volume of distribution (P = 0.84), and half-life (P = 0.36), were also not statistically significantly different. The mean area under the curve ratio between day 15 and day 1 was 3.74 in three individuals with the CYP3A5*1A/*1A genotype (all African American) compared with 1.02 in six individuals with the CYP3A5*3C/*3C genotype (all Caucasian). CONCLUSIONS: This study indicates that garlic does not significantly affect the disposition of docetaxel. However, it cannot be excluded that garlic decreases the clearance of docetaxel in patients carrying a CYP3A5*1A allele.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacocinética , Neoplasias de la Mama/tratamiento farmacológico , Ajo , Taxoides/farmacocinética , Adenocarcinoma/secundario , Administración Oral , Adulto , Anciano , Alelos , Antineoplásicos/administración & dosificación , Neoplasias de la Mama/secundario , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/genética , Docetaxel , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Estudios de Seguimiento , Genotipo , Humanos , Inyecciones Intravenosas , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Estudios Prospectivos , Taxoides/administración & dosificación , Factores de Tiempo , Resultado del Tratamiento
9.
Clin Cancer Res ; 12(13): 4004-10, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16818699

RESUMEN

PURPOSE: To determine the maximum tolerated dose, dose-limiting toxicities, and pharmacokinetic characteristics of doxorubicin encapsulated in a low temperature sensitive liposome (LTSL) when given concurrently with local hyperthermia to canine solid tumors. EXPERIMENTAL DESIGN: Privately owned dogs with solid tumors (carcinomas or sarcomas) were treated. The tumors did not involve bone and were located at sites amenable to local hyperthermia. LTSL-doxorubicin was given (0.7-1.0 mg/kg i.v.) over 30 minutes during local tumor hyperthermia in a standard phase I dose escalation study. Three treatments, given 3 weeks apart, were scheduled. Toxicity was monitored for an additional month. Pharmacokinetics were evaluated during the first treatment cycle. RESULTS: Twenty-one patients were enrolled: 18 with sarcomas and 3 with carcinomas. Grade 4 neutropenia and acute death secondary to liver failure, possibly drug related, were the dose-limiting toxicities. The maximum tolerated dose was 0.93 mg/kg. Other toxicities, with the possible exception of renal damage, were consistent with those observed following free doxorubicin administration. Of the 20 dogs that received > or = 2 doses of LTSL-doxorubicin, 12 had stable disease, and 6 had a partial response to treatment. Pharmacokinetic variables were more similar to those of free doxorubicin than the marketed liposomal product. Tumor drug concentrations at a dose of 1.0 mg/kg averaged 9.12 +/- 6.17 ng/mg tissue. CONCLUSION: LTSL-doxorubicin offers a novel approach to improving drug delivery to solid tumors. It was well tolerated and resulted in favorable response profiles in these patients. Additional evaluation in human patients is warranted.


Asunto(s)
Antineoplásicos/administración & dosificación , Carcinoma/veterinaria , Enfermedades de los Perros/terapia , Doxorrubicina/administración & dosificación , Hipertermia Inducida/métodos , Sarcoma/veterinaria , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Carcinoma/terapia , Terapia Combinada , Perros , Relación Dosis-Respuesta a Droga , Doxorrubicina/efectos adversos , Doxorrubicina/farmacocinética , Esquema de Medicación , Sistemas de Liberación de Medicamentos , Femenino , Liposomas , Masculino , Dosis Máxima Tolerada , Microondas/uso terapéutico , Sarcoma/terapia , Temperatura , Resultado del Tratamiento
10.
Cancer Manag Res ; 9: 307-314, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28744161

RESUMEN

Treatment options for patients with multiple myeloma (MM) have increased during the past decade. Despite the significant advances, challenges remain on which combination strategies will provide the optimal response for any given patient. Defining optimal combination strategies and corresponding companion diagnostics, that will guide clinical decisions are required to target relapsed or refractory multiple myeloma (RRMM) in order to improve disease progression, survival and quality of life for patients with MM. Elotuzumab is a humanized monoclonal antibody that targets signaling lymphocytic activation molecule F7 (SLAMF7), approved by the US Food and Drug Administration (FDA) in 2015 and the European Medicines Agency in 2016 for the treatment of MM. SLAMF7 is expressed in normal and malignant plasma cells and has lower expression on natural killer (NK) cells. Experimental evidence indicates that elotuzumab exhibits anti-myeloma activity through 1) antibody-dependent cell-mediated cytotoxicity, 2) enhancing NK cells cytotoxicity and 3) interfering with adhesion of MM cells to bone marrow stem cells (BMSCs). Although elotuzumab has no single agent activity in patients with RRMM who have received one to three prior therapies, the combination of elotuzumab with anti-myeloma agents, such as immunomodulatory drugs-lenalidomide, or proteasome inhibitors (PIs)-bortezomib, remarkably improved the overall response rates and progression-free survival in MM patients with only minimal incremental toxicity. In brief, the clinical data for elotuzumab indicate that targeting SLAMF7 in combination with the use of conventional therapies is feasible and effective with a tolerable safety profile for the treatment of RRMM.

11.
J Clin Oncol ; 23(25): 6117-25, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16087946

RESUMEN

PURPOSE: To evaluate associations between patient survival, pharmacokinetics, and drug metabolism-related genetic polymorphisms in patients receiving a combination chemotherapy regimen for breast cancer. PATIENTS AND METHODS: A genotype association study was conducted on 85 chemotherapy-naïve patients with metastatic or inflammatory breast cancer that were evaluated for an extended period after receiving standard-dose chemotherapy followed by high-dose cyclophosphamide, cisplatin, and carmustine. Blood pharmacokinetics were evaluated, and DNA was genotyped for 29 polymorphisms in 17 drug metabolism genes. RESULTS: Patients with cyclophosphamide plasma exposures above the median (implying slower metabolic activation) had a shorter survival than those below the median (1.8 v 3.8 years, respectively; P = .042). Patients having a variant genotype of cytochrome P450 3A4 displayed higher blood concentrations of parent (inactive) cyclophosphamide with the second and third doses (P = .024 and .028, respectively) in addition to slower cyclophosphamide activation over the three doses (P = .031). Median survival for these patients was 1.3 years compared with 2.7 years for those without the variant (P = .043). Similar results were observed for patients carrying a genetic variant of P450 3A5. Median survival for patients with deletions of glutathione-S-transferase M1 gene was 3.5 v 1.5 years for patients with one or both copies (P = .041). Patients with a polymorphism in a gene regulating metallothionein had lower platinum concentrations and shorter survival (P = .033). CONCLUSION: These data suggest that pretreatment evaluation of drug metabolism genes may explain some interindividual differences in both anticancer drug pharmacokinetics and response. The correlations found here may have implications for other commonly used anticancer drugs.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Polimorfismo Genético , Adulto , Neoplasias de la Mama/patología , Carmustina/administración & dosificación , Carmustina/farmacocinética , Cisplatino/administración & dosificación , Cisplatino/farmacocinética , Ciclofosfamida/administración & dosificación , Ciclofosfamida/farmacocinética , Femenino , Genotipo , Humanos , Persona de Mediana Edad , Pronóstico , Análisis de Supervivencia
12.
Cancer Res ; 62(12): 3408-16, 2002 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12067983

RESUMEN

Combretastatin A-4 phosphate (CA4P) is a novel antitumor vascular targeting agent, the first agent of this class of compounds to enter the clinic. We performed a Phase I trial to determine the maximum-tolerated dose, safety, and pharmacokinetic profile of CA4P on a single-dose i.v. schedule. We also obtained preliminary data on its effect on tumor blood flow using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) techniques and cell adhesion molecules at the higher-dose levels. Twenty-five assessable patients with advanced cancer received a total of 107 cycles over the following dose escalation schema: 18, 36, 60, 90 mg/m(2) as a 10-min infusion and 60 mg/m(2) as a 60-min infusion at 3-week intervals. There was no significant myelotoxicity, stomatitis, or alopecia. Tumor pain was a unique side effect, which occurred in 10% of cycles, and there were four episodes of dose-limiting toxicity at dosages > or =60 mg/m(2), including two episodes of acute coronary syndrome. Pharmacokinetics revealed rapid dephosphorylation of the parent compound (CA4P) to combretastatin A4 (CA4), with a short plasma half-life (approximately 30 min). A significant (P < 0.03) decline in gradient peak tumor blood flow by DCE-MRI in six of seven patients treated at 60 mg/m(2) was observed. A patient with anaplastic thyroid cancer had a complete response and is alive 30 months after treatment. The toxicity profile is consistent with a drug that is "vascularly active" and devoid of traditional "cytotoxic" side effects. Dosages < or =60 mg/m(2) as a 10-min infusion define the upper boundary of the maximum-tolerated dose.


Asunto(s)
Antineoplásicos Fitogénicos/efectos adversos , Antineoplásicos Fitogénicos/farmacocinética , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Estilbenos/efectos adversos , Estilbenos/farmacocinética , Adulto , Anciano , Antineoplásicos Fitogénicos/farmacología , Moléculas de Adhesión Celular/sangre , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Humanos , Infusiones Intravenosas , Angiografía por Resonancia Magnética , Masculino , Persona de Mediana Edad , Neoplasias/irrigación sanguínea , Neovascularización Patológica/metabolismo , Estilbenos/farmacología
13.
J Clin Oncol ; 21(23): 4428-38, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14645433

RESUMEN

PURPOSE: Combretastatin A4 (CA4) phosphate (CA4P) inhibits microtubule polymerization and is toxic to proliferating endothelial cells in vitro. It causes reversible vascular shutdown in established tumors in vivo, consistent with an antivascular mechanism of action. The present study investigated escalating doses of CA4P administered intravenously to patients with advanced cancer. PATIENTS AND METHODS: Patients with solid malignancies and good performance status received CA4P as a 10-minute infusion daily for 5 days repeated every 3 weeks. Pharmacokinetic sampling was performed during cycle 1. Patients receiving >/= 52 mg/m2/d had serial dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) studies to measure changes in tumor perfusion with CA4P treatment. RESULTS: Thirty-seven patients received 133 treatment cycles. CA4P dose levels ranged from 6 mg/m2 to 75 mg/m2 daily. Severe pain at sites of known tumor was dose limiting at 75 mg/m2. Dose-limiting cardiopulmonary toxicity (syncope and dyspnea or hypoxia) was noted as well in two patients treated at 75 mg/m2. Other toxicities included hypotension, ataxia, dyspnea, nausea or vomiting, headache, and transient sensory neuropathy. Plasma CA4P and CA4 area under the concentration-time curve and maximal concentration values increased linearly with dose. Tumor perfusion, as measured by the first-order rate constant of gadolinium plasma to tissue transfer during DCE-MRI studies, was found to decrease in eight of 10 patients. Relationships were also demonstrated between perfusion changes and pharmacokinetic indices. A partial response was observed in a patient with metastatic soft tissue sarcoma, and 14 patients exhibited disease stability for a minimum of two cycles. CONCLUSION: Doses of CA4P on a daily times five schedule of 52 to 65 mg/m2 were reasonably well-tolerated. The 52 mg/m2 dose is recommended for further study based on cumulative phase I experience with CA4P. Antitumor efficacy was observed, and the use of DCE-MRI provided a valuable noninvasive measure of the vascular effects of CA4P treatment.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Neoplasias/irrigación sanguínea , Neovascularización Patológica/tratamiento farmacológico , Estilbenos/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos Fitogénicos/farmacocinética , Área Bajo la Curva , Medios de Contraste , Esquema de Medicación , Femenino , Humanos , Infusiones Intravenosas , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Neoplasias/patología , Neovascularización Patológica/patología , Estilbenos/farmacocinética , Tomografía Computarizada de Emisión , Resultado del Tratamiento
14.
J Clin Oncol ; 20(3): 674-9, 2002 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11821447

RESUMEN

PURPOSE: To ascertain the maximum tolerated duration of infusion of gemcitabine at 10 mg/m(2)/min in combination with mitoxantrone at 12 mg/m(2) daily for 3 days in the treatment of acute leukemia. PATIENTS AND METHODS: Thirty-four patients were enrolled. Stratum I consisted of 26 patients, median age 50 years (range, 25 to 71 years), with relapsed or refractory leukemia. Stratum II contained eight patients, median age 62.5 years (range, 38 to 83 years), who had received fewer than three cycles of myelotoxic therapy for chronic myeloid leukemia or myelodysplasia that had evolved into leukemia. Patients received mitoxantrone at 12 mg/m(2) daily for 3 days. After the first mitoxantrone dose, gemcitabine was provided intravenously at 10 mg/m(2)/min with the duration adjusted by following a continuous reassessment model. RESULTS: Severe myelosuppression, and stomatitis or esophagitis were the most common hematologic and nonhematologic dose-limiting toxicities. Several patients developed febrile neutropenia, nausea, or vomiting. In both strata, the maximum recommended duration of infusion of gemcitabine was 12 hours (7,200 mg/m(2)). The mean steady-state concentration of gemcitabine was 24.72 micromol/L and varied over a fivefold range among patients. Overall response rates in this phase I trial for strata I and II were 42% and 63%, respectively. CONCLUSION: Prolonged-infusion gemcitabine at a fixed dose rate of 10 mg/m(2)/min for 12 hours with 12 mg/m(2)/d mitoxantrone for 3 days is a tolerable induction regimen and achieves plasma concentrations sufficient for maximal intracellular activation. Stomatitis or esophagitis should be anticipated; however, this regimen may induce significant responses in patients with difficult-to-treat leukemias.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Desoxicitidina/análogos & derivados , Desoxicitidina/administración & dosificación , Leucemia/tratamiento farmacológico , Mitoxantrona/administración & dosificación , Enfermedad Aguda , Adulto , Anciano , Antimetabolitos Antineoplásicos/farmacocinética , Antimetabolitos Antineoplásicos/toxicidad , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Desoxicitidina/farmacocinética , Desoxicitidina/toxicidad , Humanos , Infusiones Intravenosas , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Persona de Mediana Edad , Mitoxantrona/farmacocinética , Mitoxantrona/toxicidad , Defectos del Tubo Neural/tratamiento farmacológico , Gemcitabina
15.
Trends Pharmacol Sci ; 25(9): 457-64, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15559247

RESUMEN

Pharmacogenomics aims to elucidate the genomic determinants of drug disposition and effect. Because cancer chemotherapy is relatively nonspecific and has narrow therapeutic indices, there is great potential for pharmacogenomics to improve treatment outcomes by either reducing toxicity or increasing efficacy. The diversity of therapeutic targets for anticancer drugs and the intensity of clinical pharmacology research in oncology have provided many examples of clinically relevant pharmacogenomic applications. Important elements that are discussed in this article include the association of genetic variability in the metabolism, intracellular transport and targets of anticancer drugs. In addition, we summarize where the field stands currently, and how information from the host and tumor might be integrated into decision making.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Variación Genética , Neoplasias/genética , Neoplasias/metabolismo , Farmacogenética , Antineoplásicos/uso terapéutico , Humanos , Neoplasias/tratamiento farmacológico , Polimorfismo Genético
16.
Clin Cancer Res ; 8(3): 698-705, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11895898

RESUMEN

This report investigates relationships between the pharmacokinetics and pharmacodynamics of high-dose alkylators used for the treatment of primary breast cancer. Eighty-five women with primary breast cancer involving >or=10 lymph nodes received four cycles of standard-dose chemotherapy followed by a high-dose regimen consisting of: cyclophosphamide (1875 mg/m(2) once daily x 3), cisplatin (165 mg/m(2) given over 72 h), carmustine (600 mg/m(2)), and stem cell transplantation. Dosages were attenuated in patients whose body weight exceeded their calculated ideal weight by >20%. Pharmacokinetics of the high-dose chemotherapeutic agents were evaluated in each patient by collection and analysis of serial blood samples. Area under the concentration time curve (AUC) for cyclophosphamide and carmustine was highly variable (>10-fold inter-patient range) with coefficients of variation > 50%, in contrast to cisplatin exposures (2-fold range; coefficient of variation 12%). The dosing method for overweight patients resulted in significantly lower systemic exposure to cisplatin (P = 0.035). The parent cyclophosphamide clearance on the 1st day of administration was significantly higher in patients who experienced acute cardiac toxicity (n = 5; P = 0.011), whereas carmustine disposition was not found to be different in those developing pulmonary toxicity (n = 25; P = 0.96). Kaplan-Meier analysis (median follow-up of 5.9 years) demonstrated that patients with lower cyclophosphamide AUC (faster parent drug clearance to potentially cytotoxic compounds) survived longer (P = 0.031). Inter-individual differences in the pharmacokinetic disposition of high-dose chemotherapy may explain variability in both response and toxicity. Prospective strategies, which attempt to individualize AUC, should be evaluated in this setting.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Peso Corporal/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Área Bajo la Curva , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/secundario , Carmustina/administración & dosificación , Carmustina/efectos adversos , Carmustina/farmacocinética , Cisplatino/administración & dosificación , Cisplatino/efectos adversos , Cisplatino/farmacocinética , Ciclofosfamida/administración & dosificación , Ciclofosfamida/efectos adversos , Ciclofosfamida/farmacocinética , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Humanos , Metástasis Linfática , Tasa de Supervivencia , Factores de Tiempo
17.
Int J Radiat Oncol Biol Phys ; 58(3): 779-85, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14967434

RESUMEN

PURPOSE: Eniluracil, an effective inactivator of dihydropyrimidine dehydrogenase, allows for oral dosing of 5-fluorouracil (5-FU), which avoids the morbidity of continuous infusion 5-FU. We addressed the safety of oral eniluracil and 5-FU combined with preoperative radiotherapy and determined the recommended Phase II dose and dose-limiting toxicity in patients with locally advanced rectal and colon cancer. METHODS AND MATERIALS: Patients with TNM Stage II or III rectal cancer and residual or recurrent colon cancer received eniluracil (starting at 6.0 mg/m(2) every 12 h) and 5-FU (starting at 0.6 mg/m(2) every 12 h). Eniluracil and 5-FU were given with a 5-week course of preoperative radiotherapy of 4500 cGy, with a possible 540-cGy boost. Surgery was performed approximately 4 weeks after completion of chemoradiotherapy. RESULTS: Twenty-two patients were enrolled; 1 patient was withdrawn owing to noncompliance. Chemotherapy was completed in all patients; radiotherapy was completed in 20 patients. The recommended Phase II dose of eniluracil and 5-FU was 8 mg/m(2) every 12 h and 0.8 mg/m(2) every 12 h, respectively. Diarrhea was the dose-limiting toxicity. Eleven of the 17 patients with primary rectal cancer underwent a sphincter-sparing procedure. One patient had a pathologic complete response. CONCLUSION: Preoperative chemoradiotherapy with oral eniluracil and 5-FU is feasible and well tolerated. Additional investigation is warranted.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/radioterapia , Antimetabolitos Antineoplásicos/efectos adversos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/radioterapia , Inhibidores Enzimáticos/administración & dosificación , Fluorouracilo/efectos adversos , Neoplasias del Recto/tratamiento farmacológico , Neoplasias del Recto/radioterapia , Uracilo/análogos & derivados , Uracilo/efectos adversos , Adenocarcinoma/patología , Adenocarcinoma/cirugía , Administración Oral , Adulto , Anciano , Anciano de 80 o más Años , Antimetabolitos Antineoplásicos/administración & dosificación , Neoplasias del Colon/patología , Neoplasias del Colon/cirugía , Terapia Combinada , Combinación de Medicamentos , Inhibidores Enzimáticos/efectos adversos , Femenino , Fluorouracilo/administración & dosificación , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/radioterapia , Estadificación de Neoplasias , Neoplasias del Recto/patología , Neoplasias del Recto/cirugía , Uracilo/administración & dosificación
18.
Cancer Chemother Pharmacol ; 50(1): 37-45, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12111110

RESUMEN

PURPOSE: Fludarabine is a renally excreted agent that is an effective treatment for chronic lymphocytic leukemia (CLL), a disease predominantly of the elderly. We sought to determine whether age, renal function or pretreatment hematologic status predicted toxicity of fludarabine treatment for CLL. METHODS: We evaluated 192 patients with previously untreated B-cell CLL who were entered onto the fludarabine treatment arm (25 mg/m(2) daily for 5 days every 28 days) of CALGB study 9011, an intergroup study with participation from SWOG, CTG/NCI-C and ECOG. Patients were required to have serum creatinine within 1.5 times normal. Hematologic indices and infections were recorded during the first 28-day cycle of treatment. A time-to-toxicity endpoint was evaluated over the entire course of fludarabine treatment. Creatinine clearance (CrCl(est)) was estimated using serum creatinine, age and body mass index. RESULTS: The median age was 64 years (range 37-87 years) and median CrCl(est) was 62 ml/min (range 27-162 ml/min, interquartile range 52-79 ml/min). We found no association between age and incidence of hematologic toxicity or infection during the first cycle of treatment. There was a strong association between CrCl(est) and the time-to-toxicity endpoint. Patients with CrCl(est) below 80 ml/min had increased incidence of toxicity during their treatment course ( P<0.0001). Pretreatment anemia, thrombocytopenia and Rai stage were highly associated with the incidence of neutrophil toxicity and grade III/IV hematologic toxicities during the first cycle of treatment ( P<0.0001). CONCLUSIONS: Patient age was not an independent risk factor for fludarabine-related toxicity, but CrCl(est) was associated with time to toxicity.


Asunto(s)
Antineoplásicos/efectos adversos , Creatinina/sangre , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Vidarabina/efectos adversos , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Índice de Masa Corporal , Supervivencia sin Enfermedad , Femenino , Pruebas Hematológicas , Humanos , Pruebas de Función Renal , Leucemia Linfocítica Crónica de Células B/sangre , Recuento de Leucocitos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neutrófilos/efectos de los fármacos , Vidarabina/análogos & derivados
19.
Cancer Chemother Pharmacol ; 73(5): 991-7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24619498

RESUMEN

PURPOSE: This study was conducted to evaluate potential pharmacokinetic interactions between docetaxel and atrasentan as part of a phase I/II clinical trial. METHODS: Patients with prostate cancer were treated with intravenous docetaxel (60-75 mg/m(2)) every 3 weeks and oral atrasentan (10 mg) daily starting on day 3 of cycle 1 and then given continuously. The pharmacokinetics of both drugs were evaluated individually (cycle 1, day 1 for docetaxel; day 21 for atrasentan) and in combination (cycle 2, day 1 for both drugs). Pharmacogenomics of alpha-1-acid glycoprotein (AAG) were also explored. RESULTS: Paired pharmacokinetic data sets for both drugs were evaluable in 21 patients. Atrasentan was rapidly absorbed and plasma concentrations varied over a fourfold range at steady state within a typical patient. The median apparent oral clearance of atrasentan was 17.4 L/h in cycle 1 and was not affected by docetaxel administration (p = 0.9). Median systemic clearance of docetaxel was 51.1 L/h on the first cycle and significantly slower (p = 0.01) compared with that obtained during co-administration of atrasentan, 61.6 L/h. Docetaxel systemic clearance in cycle 1 was 70.0 L/h in patients homozygous for a variant allele in AAG compared with 44.5 L/h in those with at least one wild-type allele (p = 0.03). CONCLUSION: Genetic polymorphism in AAG may explain some inter-patient variability in docetaxel pharmacokinetics. The systemic clearance of docetaxel is increased by approximately 21 % when given concomitantly with atrasentan; however, atrasentan pharmacokinetics does not appear to be influenced by docetaxel administration.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Pirrolidinas/farmacología , Pirrolidinas/uso terapéutico , Taxoides/farmacología , Anciano , Anciano de 80 o más Años , Antineoplásicos/farmacocinética , Atrasentán , Docetaxel , Humanos , Masculino , Persona de Mediana Edad , Pirrolidinas/farmacocinética , Taxoides/farmacocinética , Taxoides/uso terapéutico
20.
J Hum Lact ; 29(4): 469-72, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23492761

RESUMEN

Cisplatin, a platinum-based chemotherapy agent, is commonly used in treating cancers that may affect women of childbearing age, including cervical cancer, triple-negative breast cancer, and pediatric tumors in adolescents. The authors found that platinum was undetectable in breast milk at 66 hours and beyond following a 70-mg dose of intravenous cisplatin. Relative infant dose of platinum was calculated to be between 0.29% and 0.40% of the maternal dose corrected for body weight. This case demonstrates minimal exposure to platinum via breast milk, following a single 70-mg intravenous dose of cisplatin.


Asunto(s)
Lactancia Materna , Cisplatino/análisis , Cisplatino/farmacocinética , Leche Humana/química , Adenocarcinoma/tratamiento farmacológico , Adulto , Cisplatino/uso terapéutico , Femenino , Humanos , Lactante , Factores de Tiempo , Neoplasias del Cuello Uterino/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA