Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Support Care Cancer ; 23(1): 151-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25015057

RESUMEN

PURPOSE: Because as many as 30% of cancer patients who receive chemotherapy of moderate or high emetogenic potential suffer from chemotherapy-induced nausea and vomiting (CINV), we undertook a multinational survey to identify health care providers' perceived knowledge gaps, barriers,and educational interests relevant to CINV. METHODS: An Internet-based survey was developed and was electronically disseminated to members of Medscape, an international Internet-based continuing medical education provider. RESULTS: A total of 2388 health care providers responded to the survey. Although breakthrough nausea and vomiting was the most common CINV-related issue they managed in the preceding year, managing delayed nausea was the most problematic in that time period. Thirty-two percent of health care providers delayed or discontinued a patient's chemotherapy because of CINV. Cost of antiemetics, patients' poor adherence to antiemetic regimens, and health care providers' underestimation of risk for CINV were all barriers to effective management. Health care providers expressed a wide range of educational interests, including managing breakthrough CINV, keeping up with novel antiemetic agents, and learning about emerging approaches for CINV prevention/management. CONCLUSIONS: This survey of health care providers uncovered key barriers and educational needs relevant to the management of CINV. The findings from this survey can be used to develop educational initiatives focused on improving the care of cancer patients at risk for or suffering from CINV.


Asunto(s)
Antineoplásicos/efectos adversos , Conocimientos, Actitudes y Práctica en Salud , Personal de Salud/educación , Náusea/inducido químicamente , Neoplasias/tratamiento farmacológico , Vómitos/inducido químicamente , Anciano , Antieméticos/uso terapéutico , Antineoplásicos/uso terapéutico , Recolección de Datos , Femenino , Necesidades y Demandas de Servicios de Salud , Humanos , Masculino , Persona de Mediana Edad , Náusea/tratamiento farmacológico , Percepción , Estudios Prospectivos , Vómitos/tratamiento farmacológico
2.
Bioconjug Chem ; 21(1): 20-7, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20020769

RESUMEN

Targeted delivery of therapeutic drugs promises to become the norm to treat cancer. Here, we conjugated the cytotoxic agent 6-hydroxypropylacylfulvene (HPAF) to anginex, a peptide that targets galectin-1, which is highly expressed in endothelial cells of tumor vessels. In a human ovarian cancer model in mice, the conjugate inhibited tumor growth better than equivalent doses of either compound alone. Immunofluorescence on tumor tissue demonstrated that the conjugate, like parent anginex, selectively targeted tumor vasculature and inhibited tumor angiogenesis. Increased activity from the conjugate further suggests that HPAF retains at least some of its normal cytotoxic activity when linked to anginex. More importantly perhaps is the observation that the conjugate abrogates apparent systemic toxicity from treatment with HPAF. This work contributes to the development of tumor vascular targeting agents against cancer in the clinic.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Galectina 1/antagonistas & inhibidores , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico , Proteínas/química , Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , División Celular/efectos de los fármacos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Galectina 1/metabolismo , Humanos , Neoplasias/tratamiento farmacológico , Péptidos , Proteínas/farmacología , Proteínas/uso terapéutico , Sesquiterpenos/química , Sesquiterpenos/farmacología , Sesquiterpenos/uso terapéutico , Compuestos de Espiro/química , Compuestos de Espiro/farmacología , Compuestos de Espiro/uso terapéutico , Células Tumorales Cultivadas , Venas Umbilicales/citología
4.
Clin Cancer Res ; 14(8): 2444-9, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18413836

RESUMEN

PURPOSE: Epigenetic silencing via aberrant promoter DNA hypermethylation of normal genes has been described as a leukemogenic mechanism in myelodysplastic syndromes (MDS) and acute myeloid leukemias (AML). We hypothesized that MG98, an oligonucleotide antisense to DNA methyltransferase 1 (DNMT1), could reverse malignant phenotypes by down-regulating DNMT1 and inducing reexpression of hypermethylated genes. This phase I study was conducted to determine a biologically effective dose and describe the safety of MG98 in MDS/AML. EXPERIMENTAL DESIGN: Twenty-three patients with MDS (n = 11) and AML (n = 12) were enrolled. Biologically effective dose was defined as the dose at which > or =50% of patients experienced >50% reduction in DNMT1 expression with acceptable toxicity. Escalating doses of MG98 were administered according to two schedules (2-hour i.v. bolus followed by 5-day continuous i.v. infusion every 14 days, or 14-day continuous i.v. infusion every 21 days). RESULTS: DNMT1 down-regulation was observed in 8 patients. However, biologically effective dose was not reached. Reexpression of target genes (P15, WIT1, and ER) was observed in 12 patients but did not correlate with DNMT1 down-regulation. Escalation was stopped due to dose-limiting toxicities (bone pain, nausea, and fever). No objective clinical response was observed. Disease stabilization occurred in 6 (26%) patients. CONCLUSIONS: No pharmacodynamic or clinical activity was observed at MG98 doses and schedules administered. Despite this, pursuing DNMT1 down-regulation remains a sound approach for targeting aberrant epigenetics in AML/MDS. Future studies with different formulation and/or doses and schedules will be required to ensure efficient MG98 intracellular uptake and fully evaluate its therapeutic potential.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Leucemia Mieloide Aguda/tratamiento farmacológico , Síndromes Mielodisplásicos/tratamiento farmacológico , Oligodesoxirribonucleótidos/uso terapéutico , Tionucleótidos/uso terapéutico , Anciano , Anciano de 80 o más Años , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oligodesoxirribonucleótidos/efectos adversos , Oligodesoxirribonucleótidos/farmacocinética , ARN Mensajero/análisis , Tionucleótidos/efectos adversos , Tionucleótidos/farmacocinética
5.
Cancer Lett ; 265(2): 270-80, 2008 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-18378392

RESUMEN

Combination of chemotherapeutic agents and angiogenesis inhibitors is now commonly employed in the clinic to treat cancer. Here, we used angiostatic agents anginex and 0118, in combination with the chemotherapeutic irofulven, to treat human ovarian tumor xenografts in mice. General linear mixed models were used to statistically analyze tumor growth curves. Overall, combination of a low, non-toxic dose of irofulven with either angiogenesis inhibitor was more effective at inhibiting tumor growth than any of the single agent therapies. For example, the anginex/irofulven and 0118/irofulven combinations inhibited tumor growth relative to controls by 92% (p<0.0001) and 96% (p<0.0001), respectively, with the 0118/irofulven combinations yielding 100% complete responses. This study suggests that combination therapy of 0118 or anginex and irofulven may be highly effective in the clinical setting.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Proteínas/uso terapéutico , Sesquiterpenos/uso terapéutico , Animales , Calixarenos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Ratones , Péptidos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Transl Behav Med ; 8(2): 175-182, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29390159

RESUMEN

New treatments for chronic lymphocytic leukemia (CLL) with excellent response rates and varying toxicity profiles have emerged in recent years, creating an opportunity for a patient's personal preferences to contribute to treatment decisions. We conducted a prospective, quasi-experimental pre- and post-evaluation of a multilevel educational program and its impact on knowledge of CLL and shared decision-making (SDM). We educated patients, lay navigators, nurses/advanced practice providers (APPs), and physicians. Patients were evaluated for change in patient activation, distress, desired role in decision-making, perception of decision-making, satisfaction with oncologist explanation of treatment choice, and knowledge of CLL. Lay navigators, nurses/APPs, and physicians were evaluated for change in CLL knowledge and perception of decision-making. Forty-four patients, 33 lay navigators, 27 nurses/APPs, and 27 physicians participated in the educational program. We observed trends toward improved patient activation, with 68% before education versus 76% after education reporting a Patient Activation Measure (PAM) score of 3 or 4. The percentage of patients desiring and perceiving SDM trended upward from 47% to 67% and from 35% to 49%, respectively. The percentage of patients understanding that CLL is incurable increased from 80% to 90%, as did reporting awareness of signs of progression (64% to 76%). Patients' satisfaction with their oncologists' explanations of therapy increased significantly from 83% to 95% (p = .03). CLL knowledge increased after education for lay navigators (36% vs 63%) and nurses/APPs (35% vs 69%), and remained high for physicians (85% vs 87%). Nurses/APPs and physicians perceived at least some patient involvement in decision-making at baseline, whereas 12% of patients and 23% of lay navigators perceived that physicians made decisions independently. This project demonstrated trends toward improvements in patient engagement, prognostic awareness, knowledge of signs of progression, and SDM. These promising findings should be tested in larger samples. There remains an opportunity for further improvement in SDM.


Asunto(s)
Toma de Decisiones , Leucemia Linfocítica Crónica de Células B/psicología , Leucemia Linfocítica Crónica de Células B/terapia , Educación del Paciente como Asunto , Adulto , Anciano , Femenino , Estudios de Seguimiento , Conocimientos, Actitudes y Práctica en Salud , Personal de Salud/psicología , Humanos , Masculino , Grupo de Atención al Paciente , Educación del Paciente como Asunto/métodos , Participación del Paciente/métodos , Participación del Paciente/psicología , Proyectos Piloto , Estudios Prospectivos , Estrés Psicológico
7.
Anticancer Res ; 24(1): 59-65, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15015576

RESUMEN

BACKGROUND: Irofulven (MGI 114), a novel antitumor agent synthesized from the natural product illudin S, has a unique mechanism of action involving macromolecule adduct formation, S-phase arrest and induction of apoptosis. MATERIALS AND METHODS: This study utilized MiaPaCa pancreatic xenografts to demonstrate irofulven antitumor activity using either a daily or intermittent dosing schedule. Additionally, irofulven and gemcitabine were tested in vitro and in vivo to assess the anticancer activity of the combination. RESULTS: Both dosing regimens of irofulven demonstrated curative activity against the MiaPaCa xenografts. Similar activity of irofulven on the intermittent schedule was observed at lower total doses compared to the daily dosing schedule. Furthermore, enhanced antitumor activity was observed when irofulven and gemcitabine were combined compared to single agent activity. CONCLUSION: These results support further clinical characterization of intermittent irofulven dosing schedules and suggest that irofulven combined with gemcitabine may have activity in patients with pancreatic tumors.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/tratamiento farmacológico , Sesquiterpenos/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Desoxicitidina/administración & dosificación , Esquema de Medicación , Sinergismo Farmacológico , Femenino , Humanos , Concentración 50 Inhibidora , Ratones , Ratones Desnudos , Sesquiterpenos/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
8.
Prostate ; 59(1): 22-32, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-14991863

RESUMEN

BACKGROUND: Irofulven (6-hydroxymethylacylfulvene, HMAF, MGI 114) is a novel antitumor agent currently undergoing clinical trials in hormone-refractory prostate cancer. This report examines the efficacy of irofulven alone or in combination with mitoxantrone or docetaxel against androgen-independent prostate cancer cell lines. METHODS: To elucidate the activity of irofulven monotherapy and in combination, PC-3 and DU-145 cell lines were utilized in cellular viability assessments and tumor growth inhibition studies. RESULTS: Viability assays with irofulven and mitoxantrone show additive to synergistic activity. Furthermore, irofulven and mitoxantrone in combination exhibit enhanced antitumor activity against PC-3 and DU-145 xenografts. Additive combination effects are also observed when irofulven and docetaxel were tested against PC-3 xenografts and curative activity (8/10 CR) is observed in DU-145 xenografts. CONCLUSIONS: These studies demonstrate that irofulven displays strong activity as monotherapy and in combination with mitoxantrone or docetaxel against androgen-independent prostate cancer in vitro and in vivo; thus, supporting the clinical investigation of irofulven against hormone-refractory prostate cancer.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Sesquiterpenos/farmacología , Animales , Antineoplásicos Alquilantes/administración & dosificación , Línea Celular Tumoral , Docetaxel , Sinergismo Farmacológico , Humanos , Masculino , Ratones , Ratones Desnudos , Mitoxantrona/administración & dosificación , Neoplasias de la Próstata/metabolismo , Sesquiterpenos/administración & dosificación , Taxoides/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Biol Chem ; 279(38): 39584-92, 2004 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-15269203

RESUMEN

Irofulven (6-hydroxymethylacylfulvene, HMAF, MGI 114) is one of a new class of anticancer agents that are semisynthetic derivatives of the mushroom toxin illudin S. Preclinical studies and clinical trials have demonstrated that irofulven is effective against several tumor types. Mechanisms of action studies indicate that irofulven induces DNA damage, MAPK activation, and apoptosis. In this study we found that in ovarian cancer cells, CHK2 kinase is activated by irofulven while CHK1 kinase is not activated even when treated at higher concentrations of the drug. By using GM00847 human fibroblast expressing tetracycline-controlled, FLAG-tagged kinase-dead ATR (ATR.kd), it was demonstrated that ATR kinase does not play a major role in irofulven-induced CHK2 activation. Results from human fibroblasts proficient or deficient in ATM function (GM00637 and GM05849) indicated that CHK2 activation by irofulven is mediated by the upstream ATM kinase. Phosphorylation of ATM on Ser(1981), which is critical for kinase activation, was observed in ovarian cancer cell lines treated with irofulven. RNA interference results confirmed that CHK2 activation was inhibited after introducing siRNA for ATM. Finally, experiments done with human colon cancer cell line HCT116 and its isogenic CHK2 knockout derivative; and experiments done by expressing kinase-dead CHK2 in an ovarian cancer cell line demonstrated that CHK2 activation contributes to irofulven-induced S phase arrest. In addition, it was shown that NBS1, SMC1, and p53 were phosphorylated in an ATM-dependent manner, and p53 phosphorylation on serine 20 is dependent on CHK2 after irofulven treatment. In summary, we found that the anticancer agent, irofulven, activates the ATM-CHK2 DNA damage-signaling pathway, and CHK2 activation contributes to S phase cell cycle arrest induced by irofulven.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Neoplasias Ováricas , Proteínas Serina-Treonina Quinasas/metabolismo , Sesquiterpenos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2 , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN , Femenino , Humanos , Proteínas Nucleares/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Quinasas/metabolismo , Fase S/efectos de los fármacos , Serina/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA