Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Chem Commun (Camb) ; 56(92): 14373-14376, 2020 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-33140750

RESUMEN

Novel tetraplatinated metalloporphyrin-based photosensitizers (PSs) are reported, which show excellent phototoxic indexes (PIs) up to 5800 against HeLa cells, which is, to the best of our knowledge, the highest value reported for any porphyrin so far. Furthermore, 67Zn isotope labelling allowed the determination of the ratio of zinc to platinum inside the cells using ICP-MS.


Asunto(s)
Metaloporfirinas/química , Fármacos Fotosensibilizantes/química , Platino (Metal)/química , Isótopos de Zinc/química , Cobre/química , Células HeLa , Humanos , Marcaje Isotópico , Ligandos , Espectrometría de Masas , Metaloporfirinas/efectos adversos , Imagen Óptica , Fotoquimioterapia , Fármacos Fotosensibilizantes/efectos adversos , Platino (Metal)/efectos adversos , Relación Estructura-Actividad , Distribución Tisular
2.
PLoS One ; 12(12): e0188535, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29200431

RESUMEN

Increased interest in clinical application of photodynamic therapy (PDT) in various medical fields poses a demand for better understanding of processes triggered by photo-treatment. Most of the work on PDT performed so far has focused on the immediate effects of photo-treatment. It is generally accepted that cellular damage occurs during light exposure and within a short period thereafter. If cells are not killed during the PDT, they might recover, depending on the extent of the photo-induced damage. Little is known, however, about the relationship between the properties of photosensitizers (PSs) and the delayed consequences of PDT. The aim of this work was to investigate cellular responses to sub-lethal photodynamic treatment and how toxicogenic potency may be affected by molecular features of the PS. Results demonstrated that for cationic porphyrin-based PSs, lipophilicity is the main factor determining the fate of the cells in the 24-hour post-illumination period. PSs with amphiphilic properties initiated oxidative reactions that continued in the dark, long after light exposure, and caused suppression of metabolism and loss of cell viability with concomitant changes in electrophoretic mobility of proteins, including caspases. Apoptotic activity was not stimulated in the post-illumination period. This study demonstrated that in PDT mediated by amphiphilic cationic metalloporphyrin PSs, even when immediate photo-damage is relatively mild, destructive oxidative processes initiated during PDT continue in the absence of light to substantially impair metabolism, and that post-illumination protein modification may modify utilization of cell death pathways.


Asunto(s)
Muerte Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Luz/efectos adversos , Metaloporfirinas/efectos adversos , Fotoquimioterapia/efectos adversos , Fármacos Fotosensibilizantes/efectos adversos , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Células MCF-7 , Metaloporfirinas/química , Oxidación-Reducción/efectos de los fármacos , Oxidación-Reducción/efectos de la radiación , Fármacos Fotosensibilizantes/química , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de la radiación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Tensoactivos/efectos adversos , Tensoactivos/química
3.
Int J Radiat Oncol Biol Phys ; 91(5): 961-7, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25832688

RESUMEN

PURPOSE: The purpose of phase 1 was to determine the maximum tolerated dose (MTD) of motexafin gadolinium (MGd) given concurrently with temozolomide (TMZ) and radiation therapy (RT) in patients with newly diagnosed supratentorial glioblastoma multiforme (GBM). Phase 2 determined whether this combination improved overall survival (OS) and progression-free survival (PFS) in GBM recursive partitioning analysis class III to V patients compared to therapies for recently published historical controls. METHODS AND MATERIALS: Dose escalation in phase 1 progressed through 3 cohorts until 2 of 6 patients experienced dose-limiting toxicity or a dose of 5 mg/kg was reached. Once MTD was established, a 1-sided 1-sample log-rank test at significance level of .1 had 85% power to detect a median survival difference (13.69 vs 18.48 months) with 60 deaths over a 12-month accrual period and an additional 18 months of follow-up. OS and PFS were estimated using the Kaplan-Meier method. RESULTS: In phase 1, 24 patients were enrolled. The MTD established was 5 mg/kg, given intravenously 5 days a week for the first 10 RT fractions, then 3 times a week for the duration of RT. The 7 patients enrolled in the third dose level and the 94 enrolled in phase 2 received this dose. Of these 101 patients, 87 were eligible and evaluable. Median survival time was 15.6 months (95% confidence interval [CI]: 12.9-17.6 months), not significantly different from that of the historical control (P=.36). Median PFS was 7.6 months (95% CI: 5.7-9.6 months). One patient (1%) experienced a grade 5 adverse event possibly related to therapy during the concurrent phase, and none experience toxicity during adjuvant TMZ therapy. CONCLUSIONS: Treatment was well tolerated, but median OS did not reach improvement specified by protocol compared to historical control, indicating that the combination of standard RT with TMZ and MGd did not achieve a significant survival advantage.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/terapia , Quimioradioterapia/efectos adversos , Glioblastoma/mortalidad , Glioblastoma/terapia , Neoplasias Supratentoriales/mortalidad , Neoplasias Supratentoriales/terapia , Antineoplásicos Alquilantes/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/antagonistas & inhibidores , Quimioradioterapia/métodos , Dacarbazina/administración & dosificación , Dacarbazina/efectos adversos , Dacarbazina/análogos & derivados , Supervivencia sin Enfermedad , Fraccionamiento de la Dosis de Radiación , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Metaloporfirinas/administración & dosificación , Metaloporfirinas/efectos adversos , Análisis Multivariante , Temozolomida
4.
Diabetes Metab Res Rev ; 30(8): 669-78, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24687457

RESUMEN

BACKGROUND: Peroxynitrite, a product of the reaction of superoxide with nitric oxide, causes oxidative stress with concomitant inactivation of enzymes, poly(ADP-ribosylation), mitochondrial dysfunction and impaired stress signalling, as well as protein nitration. In this study, we sought to determine the effect of preventing protein nitration or increasing peroxynitrite decomposition on diabetic neuropathy in mice after an extended period of untreated diabetes. METHODS: C57Bl6/J male control and diabetic mice were treated with the peroxynitrite decomposition catalyst Fe(III) tetramesitylporphyrin octasulfonate (FeTMPS, 10 mg/kg/day) or protein nitration inhibitor (-)-epicatechin gallate (20 mg/kg/day) for 4 weeks, after an initial 28 weeks of hyperglycaemia. RESULTS: Untreated diabetic mice developed motor and sensory nerve conduction velocity deficits, thermal and mechanical hypoalgesia, tactile allodynia and loss of intraepidermal nerve fibres. Both FeTMPS and epicatechin gallate partially corrected sensory nerve conduction slowing and small sensory nerve fibre dysfunction without alleviation of hyperglycaemia. Correction of motor nerve conduction deficit and increase in intraepidermal nerve fibre density were found with FeTMPS treatment only. CONCLUSIONS: Peroxynitrite injury and protein nitration are implicated in the development of diabetic peripheral neuropathy. The findings indicate that both structural and functional changes of chronic diabetic peripheral neuropathy can be reversed and provide rationale for the development of a new generation of antioxidants and peroxynitrite decomposition catalysts for treatment of diabetic peripheral neuropathy.


Asunto(s)
Diabetes Mellitus Tipo 1/complicaciones , Neuropatías Diabéticas/metabolismo , Epidermis/inervación , Proteínas del Tejido Nervioso/metabolismo , Sistema Nervioso Periférico/metabolismo , Ácido Peroxinitroso/metabolismo , Animales , Antioxidantes/efectos adversos , Antioxidantes/uso terapéutico , Conducta Animal/efectos de los fármacos , Catequina/efectos adversos , Catequina/análogos & derivados , Catequina/uso terapéutico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Neuropatías Diabéticas/patología , Neuropatías Diabéticas/fisiopatología , Neuropatías Diabéticas/prevención & control , Epidermis/efectos de los fármacos , Epidermis/metabolismo , Epidermis/patología , Compuestos Férricos/efectos adversos , Compuestos Férricos/uso terapéutico , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Masculino , Metaloporfirinas/efectos adversos , Metaloporfirinas/uso terapéutico , Ratones Endogámicos C57BL , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Conducción Nerviosa/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Sistema Nervioso Periférico/efectos de los fármacos , Sistema Nervioso Periférico/patología , Sistema Nervioso Periférico/fisiopatología , Ácido Peroxinitroso/antagonistas & inhibidores , Tiempo de Reacción/efectos de los fármacos , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/patología
5.
Diabetes Obes Metab ; 15(11): 1029-39, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23731386

RESUMEN

AIM: Visceral adiposity and impaired glucose metabolism are common patho-physiological features in patients co-morbid with obesity and type-2 diabetes. We investigated the effects of the heme-oxygenase (HO) inducer hemin and the HO blocker stannous-mesoporphyrin (SnMP) on glucose metabolism, adipocyte hypertrophy and pro-inflammatory cytokines/mediators in Zucker diabetic fatty (ZDF) rats, a model characterized by obesity and type-2 diabetes. METHODS: Histological, morphological/morphometrical, Western immunoblotting, enzyme immunoassay, ELISA and spectrophotometric analysis were used. RESULTS: Treatment with hemin enhanced HO-1, HO activity and cGMP, but suppressed retroperitoneal adiposity and abated the elevated levels of macrophage-chemoattractant protein-1 (MCP-1), ICAM-1, tumour necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), IL-1ß, NF-κB, c-Jun-NH2-terminal-kinase (JNK) and activating-protein (AP-1), with parallel reduction of adipocyte hypertrophy. Correspondingly, important proteins of lipid metabolism and insulin-signalling such as lipoprotein lipase (LPL), insulin-receptor substrate-1 (IRS-1), GLUT4, PKB/Akt, adiponectin, the insulin-sensitizing and anti-inflammatory protein and adenosine-monophosphate-activated protein kinase (AMPK) were significantly enhanced in hemin-treated ZDF rats. CONCLUSION: Elevated retroperitoneal adiposity and the high levels of MCP-1, ICAM-1, TNF-α, IL-6, IL-1ß, NF-κB, JNK and AP-1 in untreated ZDF are patho-physiological factors that exacerbate inflammatory insults, aggravate adipocyte hypertrophy, with corresponding reduction of adiponectin and deregulation of insulin-signalling and lipid metabolism. Therefore, the suppression of MCP-1, ICAM-1, TNF-α, IL-6, IL-1ß, NF-κB, JNK, AP-1 and adipocyte hypertrophy, with the associated enhancement of LPL, adiponectin, AMPK, IRS-1, GLUT4, PKB/Akt and cGMP in hemin-treated ZDF are among the multifaceted mechanisms by which the HO system combats inflammation to potentiate insulin signalling and improve glucose and lipid metabolism. Thus, HO inducers may be explored in the search of novel remedies against the co-morbidities of obesity, dysfunctional lipid metabolism and impaired glucose metabolism.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hemina/uso terapéutico , Hipoglucemiantes/uso terapéutico , Resistencia a la Insulina , Grasa Intraabdominal/efectos de los fármacos , Obesidad/complicaciones , Adiponectina/metabolismo , Adiposidad/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/antagonistas & inhibidores , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/metabolismo , Inducción Enzimática/efectos de los fármacos , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/biosíntesis , Hemo-Oxigenasa 1/metabolismo , Hemina/antagonistas & inhibidores , Hiperglucemia/prevención & control , Hipertrofia , Hipoglucemiantes/antagonistas & inhibidores , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Grasa Intraabdominal/inmunología , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/patología , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Metaloporfirinas/efectos adversos , Distribución Aleatoria , Ratas , Ratas Zucker , Transducción de Señal/efectos de los fármacos
6.
Int J Radiat Oncol Biol Phys ; 85(1): e55-60, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23092726

RESUMEN

PURPOSE: To evaluate the effects on 1-year event-free survival (EFS) and overall survival (OS) of combining motexafin and gadolinium (MGd), a potent radiosensitizer, with daily fractionated radiation therapy in children with newly diagnosed intrinsic pontine gliomas. METHODS AND MATERIALS: Patients with newly diagnosed intrinsic pontine glioma were treated with MGd daily for 5 consecutive days each week, for a total of 30 doses. Patients received a 5- to 10-min intravenous bolus of MGd, 4.4 mg/kg/day, given 2 to 5 h prior to standard dose irradiation. Radiation therapy was administered at a daily dose of 1.8 Gy for 30 treatments over 6 weeks. The total dose was 54 Gy. RESULTS: Sixty eligible children received MGd daily, concurrent with 6 weeks of radiation therapy. The estimated 1-year EFS was 18%±5%, and the estimated 1-year OS was 53%±6.5%. The most common grade 3 to 4 toxicities were lymphopenia, transient elevation of liver transaminases, and hypertension. CONCLUSIONS: Compared to historical controls, the addition of MGd to a standard 6-week course of radiation did not improve the survival of pediatric patients with newly diagnosed intrinsic pontine gliomas.


Asunto(s)
Neoplasias del Tronco Encefálico/radioterapia , Glioma/radioterapia , Metaloporfirinas/uso terapéutico , Puente , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Adolescente , Neoplasias del Tronco Encefálico/mortalidad , Niño , Preescolar , Supervivencia sin Enfermedad , Fraccionamiento de la Dosis de Radiación , Esquema de Medicación , Femenino , Glioma/mortalidad , Humanos , Hipertensión/inducido químicamente , Lactante , Infusiones Intravenosas , Hígado/enzimología , Linfopenia/inducido químicamente , Masculino , Metaloporfirinas/efectos adversos , Fármacos Sensibilizantes a Radiaciones/efectos adversos
7.
Invest New Drugs ; 29(2): 316-22, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19997959

RESUMEN

PURPOSE: To assess the safety, maximum-tolerated dose (MTD), and dose-limiting toxicities (DLT), of motexafin gadolinium (MGd), given in combination with doxorubicin, in patients with advanced solid tumors. STUDY DESIGN: The combination of MGd and doxorubicin was administered every 28 days (cycle 1) and then every 21 days (subsequent cycles). The dose of MGd, given daily for 3 days, was escalated from 1.0 mg/kg/d to 3.3 mg/kg/d, while the dose of doxorubicin was held at 30 mg/m². RESULTS: Fifteen patients received 37 cycles of treatment, for a median of 2 cycles per patient (range 0-6 cycles). Three patients (20%) completed 6 cycles of therapy. The MTD was identified as MGd, 2 mg/kg/day and doxorubicin, 30 mg/m². Dose limiting toxicities included grade 3 hypertension, pneumonia, bacteremia, and elevated GGT. Serious adverse events also included pulmonary embolism and urinary tract infection requiring hospitalization. There was no exacerbation of cardiac toxicity. No patients attained a response to treatment. Six patients (54%) had stable disease. The median time to disease progression, or to last assessment, was 49 days (range 8-195 days). CONCLUSIONS: The combination of MGd and doxorubicin was fairly well tolerated. However, due to emerging preclinical data suggesting that MGd inhibits ribonucleotide reductase, further development of the combination of MGd plus doxorubicin is not recommended.


Asunto(s)
Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Doxorrubicina/uso terapéutico , Metaloporfirinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Adulto , Anciano , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Demografía , Relación Dosis-Respuesta a Droga , Doxorrubicina/administración & dosificación , Doxorrubicina/efectos adversos , Femenino , Humanos , Masculino , Metaloporfirinas/administración & dosificación , Metaloporfirinas/efectos adversos , Persona de Mediana Edad , Neoplasias/patología , Resultado del Tratamiento
8.
Clin Cancer Res ; 15(20): 6462-71, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19825958

RESUMEN

PURPOSE: Therapeutic strategies to enhance the efficacy of radioimmunotherapy have not been explored. Motexafin gadolinium is a novel anticancer agent that targets redox-dependent pathways and enhances sensitivity of tumor cells to ionizing radiation. EXPERIMENTAL DESIGN: We did preclinical studies examining motexafin gadolinium combined with rituximab and/or radiation in lymphoma cells. We subsequently completed a phase I clinical trial combining escalating doses of motexafin gadolinium concurrently with standard [(90)Y]ibritumomab tiuxetan for patients with relapsed/refractory non-Hodgkin's lymphoma. RESULTS: In HF1 lymphoma cells, motexafin gadolinium and rituximab resulted in synergistic cytotoxicity (combination index, 0.757) through a mitochondrial-mediated caspase-dependent pathway, whereas cell death in Ramos and SUDHL4 cells was additive. Motexafin gadolinium/rituximab combined with radiation (1-3 Gy) resulted in additive apoptosis. Twenty-eight of 30 patients were evaluable on the phase I clinical trial. Median age was 65 years (47-87 years), and histologies were marginal-zone (n = 1), mantle-cell (n = 3), diffuse large cell (n = 6), and follicular lymphoma (n = 18). Of all patients, 86% were rituximab refractory. Therapy was well tolerated, and no dose-limiting toxicity was seen. Overall response rate was 57% [complete remission (CR), 43%], with median time-to-treatment failure of 10 months (1-48+ months) and median duration-of-response of 17 months. Of note, all responses were documented at 4 weeks. Furthermore, in rituximab-refractory follicular lymphoma (n = 14), overall response rate was 86% (CR, 64%), with a median time-to-treatment failure of 14 months (2-48+ months). CONCLUSIONS: This represents the first report of a novel agent to be combined safely concurrently with radioimmunotherapy. Furthermore, tumor responses with [(90)Y]ibritumomab tiuxetan/motexafin gadolinium were prompt with a high rate of CRs, especially in rituximab-refractory follicular lymphoma.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Linfoma no Hodgkin/tratamiento farmacológico , Linfoma no Hodgkin/radioterapia , Metaloporfirinas/administración & dosificación , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Radioisótopos de Itrio/administración & dosificación , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Línea Celular Tumoral , Terapia Combinada , Esquema de Medicación , Femenino , Humanos , Masculino , Metaloporfirinas/efectos adversos , Persona de Mediana Edad , Porfirinas , Radioinmunoterapia , Recurrencia , Resultado del Tratamiento
9.
Leuk Lymphoma ; 50(12): 1977-82, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19860624

RESUMEN

Chronic lymphocytic leukemia (CLL) cells are susceptible to oxidative stress. The expanded porphyrin, motexafin gadolinium (MGd), reacts with intracellular reducing metabolites and protein thiols to generate reactive oxygen species (ROS). A phase II trial administered MGd 5 mg/kg/day IV for 5 days every 3 weeks until disease progression to patients with previously treated CLL and small lymphocytic lymphoma. Thirteen patients (median age 66 years) with a median of four prior therapies (range 2-9) were enrolled. Modest anti-tumor activity was seen in three patients, with improvement in lymphocytosis, lymphadenopathy and/or splenomegaly, but no patient achieved a partial or complete response by NCI 96 criteria. Flow cytometry confirmed tumor uptake of MGd. Serial increase in AKT phosphorylation in patient samples following MGd treatment was not observed, suggesting intracellular generation of ROS was not optimal. Therefore, this schedule of administration achieved MGd uptake into primary tumor cells in vivo, but clinical activity was modest.


Asunto(s)
Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Metaloporfirinas/uso terapéutico , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Deleción Cromosómica , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 17/genética , Esquema de Medicación , Resistencia a Antineoplásicos , Femenino , Citometría de Flujo , Humanos , Immunoblotting , Hibridación Fluorescente in Situ , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/metabolismo , Masculino , Metaloporfirinas/efectos adversos , Metaloporfirinas/farmacocinética , Persona de Mediana Edad , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Resultado del Tratamiento
10.
Cancer ; 115(19): 4459-69, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19572413

RESUMEN

BACKGROUND: Gold (III) compounds have exhibited favorable antitumor properties both in vitro and in vivo. In a previous study, the authors reported that the novel gold (III) complex 1a (gold 1a) exhibited strong cytotoxicity in some tumor cell lines. In the current study, the effect of gold 1a was investigated on colon cancer cells. METHODS: The cytotoxicity of gold 1a was determined by using the 3-(4,5-dimethyl-2-thihazyl)-2,5-diphenyl-2H-tetrazolium bromide method. Flow cytometry was used to detect apoptosis and cell cycle. The expression of protein was evaluated by Western blot assay. Tumor growth in vivo was evaluated in nude mice. RESULTS: Gold 1a exhibited marked cytotoxic effects in vitro to human colon cancer, and the concentration of drug required to inhibit cell growth by 50% compared with control (IC(50)) values ranged from 0.2 muM to 3.4 muM, which represented 8.7-fold to 20.8-fold greater potency than that of cisplatin. Gold 1a significantly induced apoptosis and cell cycle arrest and cleaved caspase 3, caspase 7, and poly(ADP-ribose) polymerase; released cytochrome C, and up-regulated p53, p21, p27, and Bax. In vivo, intraperitoneal injection of gold 1a at doses of 1.5 mg/kg and 3.0 mg/kg significantly inhibited tumor cell proliferation, induced apoptosis, and suppressed colon cancer tumor growth. An acute toxicology study indicated that gold 1a at effective antitumor concentrations did not cause any toxic side effects in mice. CONCLUSIONS: The current results suggested that gold 1a may be a new potential therapeutic drug for colon cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , Metaloporfirinas/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Metaloporfirinas/efectos adversos , Metaloporfirinas/farmacología , Ratones , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Int J Radiat Oncol Biol Phys ; 73(4): 1069-76, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18977094

RESUMEN

PURPOSE: To determine the efficacy of motexafin gadolinium (MGd) in combination with whole brain radiotherapy (WBRT) for the treatment of brain metastases from non-small-cell lung cancer. METHODS AND MATERIALS: In an international, randomized, Phase III study, patients with brain metastases from non-small-cell lung cancer were randomized to WBRT with or without MGd. The primary endpoint was the interval to neurologic progression, determined by a centralized Events Review Committee who was unaware of the treatment the patients had received. RESULTS: Of 554 patients, 275 were randomized to WBRT and 279 to WBRT+MGd. Treatment with MGd was well tolerated, and 92% of the intended doses were administered. The most common MGd-related Grade 3+ adverse events included liver function abnormalities (5.5%), asthenia (4.0%), and hypertension (4%). MGd improved the interval to neurologic progression compared with WBRT alone (15 vs. 10 months; p = 0.12, hazard ratio [HR] = 0.78) and the interval to neurocognitive progression (p = 0.057, HR = 0.78). The WBRT patients required more salvage brain surgery or radiosurgery than did the WBRT+MGd patients (54 vs. 25 salvage procedures, p < 0.001). A statistically significant interaction between the geographic region and MGd treatment effect (which was in the prespecified analysis plan) and between treatment delay and MGd treatment effect was found. In North American patients, where treatment was more prompt, a statistically significant prolongation of the interval to neurologic progression, from 8.8 months for WBRT to 24.2 months for WBRT+MGd (p = 0.004, HR = 0.53), and the interval to neurocognitive progression (p = 0.06, HR = 0.73) were observed. CONCLUSION: In the intent-to-treat analysis, MGd exhibited a favorable trend in neurologic outcomes. MGd significantly prolonged the interval to neurologic progression in non-small-cell lung cancer patients with brain metastases receiving prompt WBRT. The toxicity was acceptable.


Asunto(s)
Neoplasias Encefálicas/terapia , Carcinoma de Pulmón de Células no Pequeñas/terapia , Irradiación Craneana/métodos , Neoplasias Pulmonares , Metaloporfirinas/uso terapéutico , Anciano , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/secundario , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Carcinoma de Pulmón de Células no Pequeñas/secundario , Trastornos del Conocimiento/diagnóstico , Trastornos del Conocimiento/etiología , Terapia Combinada/métodos , Femenino , Humanos , Metaloporfirinas/efectos adversos , Persona de Mediana Edad , Modelos de Riesgos Proporcionales
13.
Clin Genitourin Cancer ; 6(2): 73-8, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18824428

RESUMEN

BACKGROUND: Thioredoxin reductase (Trx) has been implicated in activation of hypoxia-inducible factor-1alpha, which is overexpressed in > 85% of renal cell carcinomas (RCCs). We evaluated the safety and efficacy of motexafin gadolinium (MGd), a Trx inhibitor, as a single-agent therapy for metastatic RCC. PATIENTS AND METHODS: Patients with metastatic RCC were infused daily with MGd 5 mg/kg on days 1-5 and days 15-19 of each 28-day cycle. Patients were evaluated for response on days 21-28 of every third cycle. Those with tumor response or stable disease (SD) continued treatment for < or = 12 cycles. Twenty-five patients with confirmed metastatic RCC were enrolled. All were evaluable for toxicity, and 20 were evaluable for response. RESULTS: While no clinical responses were observed, 8 patients had SD after 3 treatment cycles, as did 4 after 6 cycles. Median overall survival was 10.1 months, and median progression- free survival was 2.7 months. The most common treatment-related toxicities were grade 1/2 pain, nausea, skin discoloration, fatigue, blisters, and headache. The most common grade 3 toxicity was hypophosphatemia, observed in 5 patients. MGd was reasonably tolerated, and disease stabilization was observed in several patients with metastatic RCC. CONCLUSION: These results show promise for the use of MGd in combination with other molecularly targeted therapies in previously treated patients with metastatic RCC. However, further investigation of MGd alone for metastatic RCC is not recommended.


Asunto(s)
Carcinoma de Células Renales/tratamiento farmacológico , Neoplasias Renales/tratamiento farmacológico , Metaloporfirinas/administración & dosificación , Adulto , Anciano , Carcinoma de Células Renales/patología , Esquema de Medicación , Femenino , Humanos , Neoplasias Renales/patología , Masculino , Metaloporfirinas/efectos adversos , Persona de Mediana Edad , Metástasis de la Neoplasia
14.
Neuro Oncol ; 10(5): 752-8, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18715950

RESUMEN

The purpose of this study was to determine the dose-limiting toxicities, maximum tolerated dose, pharmacokinetics, and intratumor and brain distribution of motexafin gadolinium (MGd) with involved field radiation therapy in children with newly diagnosed intrinsic pontine gliomas. MGd was administered as a 5-min intravenous bolus 2-5 h prior to standard radiation. The starting dose was 1.7 mg/kg. After first establishing that 5 doses/week for 6 weeks was tolerable, the dose of MGd was escalated until dose-limiting toxicity was reached. Radiation therapy was administered to 54 Gy in 30 once-daily fractions. Forty-four children received MGd at doses of 1.7 to 9.2 mg/kg daily prior to radiation therapy for 6 weeks. The maximum tolerated dose was 4.4 mg/kg. The primary dose-limiting toxicities were grade 3 and 4 hypertension and elevations in serum transaminases. Median elimination half-life and clearance values were 6.6 h and 25.4 ml/kg/h, respectively. The estimated median survival was 313 days (95% confidence interval, 248-389 days). The maximum tolerated dose of MGd and the recommended phase II dose was 4.4 mg/kg when administered as a daily intravenous bolus in conjunction with 6 weeks of involved field radiation therapy for pediatric intrinsic pontine gliomas.


Asunto(s)
Neoplasias del Tronco Encefálico/terapia , Glioma/terapia , Metaloporfirinas/administración & dosificación , Puente/patología , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Adolescente , Adulto , Área Bajo la Curva , Niño , Preescolar , Terapia Combinada , Femenino , Semivida , Humanos , Imagen por Resonancia Magnética , Masculino , Dosis Máxima Tolerada , Metaloporfirinas/efectos adversos , Metaloporfirinas/farmacocinética , Fármacos Sensibilizantes a Radiaciones/efectos adversos , Fármacos Sensibilizantes a Radiaciones/farmacocinética , Radioterapia
15.
Int J Radiat Oncol Biol Phys ; 69(3): 831-8, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17560737

RESUMEN

PURPOSE: Motexafin gadolinium (MGd) is a putative radiation enhancer initially evaluated in patients with brain metastases. This Phase I trial studied the safety and tolerability of a 2-6-week course (10-22 doses) of MGd with radiotherapy for glioblastoma multiforme. METHODS AND MATERIALS: A total of 33 glioblastoma multiforme patients received one of seven MGd regimens starting at 10 doses of 4 mg/kg/d MGd and escalating to 22 doses of 5.3 mg/kg/d MGd (5 or 10 daily doses then three times per week). The National Cancer Institute Cancer Therapy Evaluation Program toxicity and stopping rules were applied. RESULTS: The maximal tolerated dose was 5.0 mg/kg/d MGd (5 d/wk for 2 weeks, then three times per week) for 22 doses. The dose-limiting toxicity was reversible transaminase elevation. Adverse reactions included rash/pruritus (45%), chills/fever (30%), and self-limiting vesiculobullous rash of the thumb and fingers (42%). The median survival of 17.6 months prompted a case-matched analysis. In the case-matched analysis, the MGd patients had a median survival of 16.1 months (n = 31) compared with the matched Radiation Therapy Oncology Group database patients with a median survival of 11.8 months (hazard ratio, 0.43; 95% confidence interval, 0.20-0.94). CONCLUSION: The maximal tolerated dose of MGd with radiotherapy for glioblastoma multiforme in this study was 5 mg/kg/d for 22 doses (daily for 2 weeks, then three times weekly). The baseline survival calculations suggest progression to Phase II trials is appropriate, with the addition of MGd to radiotherapy with concurrent and adjuvant temozolomide.


Asunto(s)
Neoplasias Encefálicas/radioterapia , Glioblastoma/radioterapia , Metaloporfirinas/administración & dosificación , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Adulto , Anciano , Neoplasias Encefálicas/patología , Esquema de Medicación , Femenino , Glioblastoma/patología , Humanos , Imagen por Resonancia Magnética , Masculino , Análisis por Apareamiento , Dosis Máxima Tolerada , Metaloporfirinas/efectos adversos , Persona de Mediana Edad , Fármacos Sensibilizantes a Radiaciones/efectos adversos
16.
J Environ Pathol Toxicol Oncol ; 25(1-2): 373-87, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16566729

RESUMEN

Locally recurrent prostate cancer after treatment with radiation therapy is a clinical problem with few acceptable treatments. One potential treatment, photodynamic therapy (PDT), is a modality that uses laser light, drug photosensitizer, and oxygen to kill tumor cells through direct cellular cytotoxicity and/or through destruction of tumor vasculature. A Phase I trial of interstitial PDT with the photosensitizer Motexafin lutetium was initiated in men with locally recurrent prostate cancer. In this ongoing trial, the primary objective is to determine the maximally tolerated dose of Motexafin lutetium-mediated PDT. Other objectives include evaluation of Motexafin lutetium uptake from prostate tissue using a spectrofluorometric assay and evaluation of optical properties in the human prostate. Fifteen men with biopsy-proven locally recurrent prostate cancer and no evidence of distant metastatic disease have been enrolled and 14 have been treated. Treatment plans were developed using transrectal ultrasound images. The PDT dose was escalated by increasing the Motexafin lutetium dose, increasing the 732 ran light dose, and decreasing the drug-light interval. Motexafin lutetium doses ranged from 0.5 to 2 mg/kg administered IV 24, 6, or 3 hr prior to 732 ran light delivery. The light dose, measured in real time with in situ spherical detectors was 25-100 J/cm2. Light was delivered via optical fibers inserted through a transperineal brachytherapy template in the operating room. Optical property measurements were made before and after light therapy. Prostate biopsies were obtained before and after light delivery for spectrofluorometric measurements of photosensitizer uptake. Fourteen patients have completed protocol treatment on eight dose levels without dose-limiting toxicity. Grade I genitourinary symptoms that are PDT related have been observed. One patient had Grade II urinary urgency that was urinary catheter related. No rectal or other gastrointestinal PDT-related tox-icities have been observed to date. Measurements of Motexafin lutetium demonstrated the presence of photosensitizer in prostate tissue from all patients. Optical property measurements demonstrated substantial heterogeneity in the optical properties of the human prostate gland which supports the use of individualized treatment planning for prostate PDT.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Metaloporfirinas/uso terapéutico , Fotoquimioterapia/efectos adversos , Fármacos Fotosensibilizantes/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Anciano , Humanos , Masculino , Dosis Máxima Tolerada , Metaloporfirinas/efectos adversos , Persona de Mediana Edad , Fármacos Fotosensibilizantes/efectos adversos
17.
Int J Radiat Oncol Biol Phys ; 64(2): 343-54, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16414370

RESUMEN

PURPOSE: Radiation therapy plays a critical role in the local and regional control of malignant tumors. Its efficacy, however, is limited by a number of factors, including toxicity, tumor hypoxia, and tumor genetics. Recent attempts to enhance the efficacy of radiation therapy have focused on biologic agents that modulate reduction/oxidation reactions within tumor cells. METHODS AND MATERIALS: We review five promising redox modulators that are in development. Tirapazamine and AQ4N are known as "hypoxic cell sensitizers" and are toxic in areas of low oxygen tension. RSR13 facilitates delivery of oxygen to tumor cells, thereby rendering them more sensitive to radiation. Motexafin gadolinium, with a porphyrin-like structure, selectively accumulates in tumor cells and thereby enhances radiation-induced DNA damage. HIF-1 inhibitors target a transcription factor that regulates hypoxia-related events and cell survival. RESULTS: Our review of each agent included a thorough search of published preclinical and clinical data, including that presented in abstracts and posters at international meetings. Our objectives were not to identify a superior mechanism or drug, but rather to summarize the available safety and efficacy data. CONCLUSION: Clearly, there is an unmet need for safer agents that augment the efficacy of radiation therapy. This review highlights five promising redox modulators that are in development. None has yet been approved by the Food and Drug Administration. These drugs were selected for discussion because they exemplify the current investigative landscape of radiosensitizers and are indicative of future directions in this area. These radiation sensitizers have the potential to succeed where others have failed, by locally increasing the radiosensitivity of tumor cells without enhancing that of surrounding normal tissues.


Asunto(s)
Neoplasias/radioterapia , Oxidación-Reducción/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Compuestos de Anilina/efectos adversos , Compuestos de Anilina/metabolismo , Compuestos de Anilina/uso terapéutico , Antraquinonas/efectos adversos , Antraquinonas/metabolismo , Antraquinonas/uso terapéutico , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Ensayos Clínicos como Asunto , Daño del ADN , Aprobación de Drogas , Quimioterapia Combinada , Humanos , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Factor 1 Inducible por Hipoxia/metabolismo , Dosis Máxima Tolerada , Metaloporfirinas/efectos adversos , Metaloporfirinas/metabolismo , Metaloporfirinas/uso terapéutico , Neoplasias/metabolismo , Propionatos/efectos adversos , Propionatos/metabolismo , Propionatos/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/efectos adversos , Fármacos Sensibilizantes a Radiaciones/metabolismo , Tirapazamina , Triazinas/efectos adversos , Triazinas/metabolismo , Triazinas/uso terapéutico
18.
Curr Opin Investig Drugs ; 7(1): 70-80, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16425674

RESUMEN

AEOL-10150, a small-molecule antioxidant analogous to the catalytic site of superoxide dismutase, is under development by Aeolus (formerly Incara) as a potential subcutaneous treatment for amyotrophic lateral sclerosis (ALS), stroke, spinal cord injury, lung inflammation and mucositis. The compound is currently undergoing a phase I clinical trial for ALS. In October 2005, the company had applied for Fast Track status, and planned to submit a special protocol assessment for a pivotal phase II/III trial.


Asunto(s)
Antioxidantes/uso terapéutico , Metaloporfirinas/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Antioxidantes/efectos adversos , Antioxidantes/química , Antioxidantes/farmacocinética , Ensayos Clínicos como Asunto , Humanos , Enfermedades Pulmonares/tratamiento farmacológico , Metaloporfirinas/efectos adversos , Metaloporfirinas/química , Metaloporfirinas/farmacología , Estructura Molecular , Neoplasias/tratamiento farmacológico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Fármacos Neuroprotectores/efectos adversos , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacología , Relación Estructura-Actividad , Resultado del Tratamiento
19.
Cancer Chemother Pharmacol ; 57(4): 465-74, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16133531

RESUMEN

PURPOSE: To determine the maximum tolerated dose and dose-limiting toxicity (DLT) of the novel anticancer agent, motexafin gadolinium (MGd), administered concurrently with radiation therapy (RT) in patients with locally advanced pancreatic or biliary tumors. The pharmacokinetics of MGd were also evaluated. METHODS: Cohorts of three to six patients were treated with escalating doses of MGd, administered three times per week for a total of 16 doses concurrent with RT. The dose of RT was fixed at 5,040 cGy, and given in 28 fractions, from Monday to Friday of every week. Plasma MGd concentrations were measured by high performance liquid chromatography. RESULTS: Eight patients were treated at dose level 1 (2.9 mg/kg), with one DLT (grade 3 fever). Three patients were treated at dose level 2 (3.6 mg/kg), and two DLTs were noted. One DLT was grade 3 nausea and vomiting (N/V), and the other was grade 3 skin toxicity. The most common toxicity was N/V. There were no objective responses. The median survival was 6 months. The MGd plasma concentration versus time profile in each patient was best fit by a two-compartment, open, linear model. There was minimal accumulation of MGd in plasma with the three-times/week dosing schedule. Simulation of the time course of MGd in the peripheral compartment indicated that maximal MGd concentrations of 1-2 micromol/kg occurred between 4 and 6 h after MGd infusion. CONCLUSION: Dose level 1 (2.9 mg/kg of MGd) is the recommended dose for combination with (RT) in phase II studies for locally advanced pancreatic and biliary cancers. Patient tolerance might be improved by modification of the RT schedule and antiemetic prophylaxis.


Asunto(s)
Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Neoplasias del Sistema Biliar/terapia , Metaloporfirinas/farmacocinética , Metaloporfirinas/uso terapéutico , Neoplasias Pancreáticas/terapia , Anciano , Área Bajo la Curva , Neoplasias del Sistema Biliar/tratamiento farmacológico , Neoplasias del Sistema Biliar/radioterapia , Terapia Combinada , Relación Dosis-Respuesta a Droga , Femenino , Semivida , Humanos , Pruebas de Función Hepática , Masculino , Metaloporfirinas/efectos adversos , Persona de Mediana Edad , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/radioterapia
20.
Curr Opin Pediatr ; 17(2): 167-9, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15800405

RESUMEN

PURPOSE OF REVIEW: To evaluate the safety and efficacy of metalloporphyrins for the treatment of neonatal hyperbilirubinemia. RECENT FINDING: Since the 1980s and 1990s, there have been no publications on trials to determine the efficacy of metalloporphyrins in the treatment of neonatal jaundice. In the past year, a single case report was presented on the compassionate use of tin mesoporphyrin in a very low birth weight infant with intrauterine growth retardation who did not respond to phototherapy. Subcutaneous administration of a single dose of tin mesoporphyrin at 46 hours of life was associated with a greater than 25% reduction in serum bilirubin. This further supports existing evidence that tin mesoporphyrin is efficacious in lowering bilirubin production. In the laboratory, most metalloporphyrins were shown to induce heme oxygenase, and in addition, metalloporphyrins modulate cardiac cell function in vitro. These observations suggest that the therapeutic benefits may be obviated if such considerations hold true in humans. SUMMARY: Recent case reports and previous evidence from larger clinical trials conducted in Greece and Argentina in the 1980s and 1990s demonstrate that tin mesoporphyrin is useful in the treatment of neonatal jaundice. Its long-term safety is not well understood but will be important to determine, before its widespread or prophylactic use in neonates with hyperbilirubinemia can be recommended.


Asunto(s)
Ictericia Neonatal/tratamiento farmacológico , Metaloporfirinas/uso terapéutico , Bilirrubina/sangre , Bilirrubina/metabolismo , Humanos , Recién Nacido , Metaloporfirinas/efectos adversos , Metaloporfirinas/química , Estructura Molecular , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...