Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
2.
Intern Med J ; 50(6): 716-725, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31318119

RESUMEN

BACKGROUND: Lung cancer is a major cause of death in New Zealand. In recent years, targeted therapies have improved outcomes. AIM: To determine the uptake of anaplastic lymphoma kinase (ALK) testing, and the prevalence, demographic profile and outcomes of ALK-positive non-small-cell lung cancer (NSCLC), in New Zealand, where no national ALK-testing guidelines or subsidised ALK tyrosine kinase inhibitor (TKI) therapies are available. METHODS: A population-based observational study reviewed databases to identify patients presenting with non-squamous NSCLC over 6.5 years in northern New Zealand. We report the proportion tested for ALK gene rearrangements and the results. NSCLC samples tested by fluorescence in situ hybridisation were retested by next generation sequencing and ALK immunohistochemistry. A survival analysis compared ALK-positive patients treated or not treated with ALK TKI therapy. RESULTS: From a total of 3130 patients diagnosed with non-squamous NSCLC, 407 (13%) were tested for ALK gene rearrangements, and patient selection was variable and inequitable. Among those tested, 34 (8.4%) had ALK-positive NSCLC. ALK-positive disease was more prevalent in younger versus older patients, non-smokers versus smokers and in Maori, Pacific or Asian ethnic groups than in New Zealand Europeans. Fluorescence in situ hybridisation, ALK immunohistochemistry and next generation sequencing showed broad concordance for detecting ALK-positive disease under local testing conditions. Among patients with ALK-positive metastatic NSCLC, those treated with ALK TKI survived markedly longer than those not treated with ALK TKI (median overall survival 5.12 vs 0.55 years). CONCLUSION: Lung cancer outcomes in New Zealand may be improved by providing national guidelines and funding policy for ALK testing and access to subsidised ALK TKI therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Quinasa de Linfoma Anaplásico/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Detección Precoz del Cáncer , Reordenamiento Génico , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/epidemiología , Nueva Zelanda/epidemiología , Inhibidores de Proteínas Quinasas , Proteínas Tirosina Quinasas Receptoras/genética
3.
Cancer Epidemiol ; 57: 24-32, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30278336

RESUMEN

BACKGROUND: Epidermal Growth Factor Receptor (EGFR) mutation testing is recommended for patients with non-squamous non-small cell lung cancer (NSCLC) but not all eligible patients get tested, which may bias the mutation prevalence estimated. This study aims to examine trends in the uptake of EGFR mutation testing in patients with non-squamous NSCLC in New Zealand; to develop a composite metric that quantifies the influences of demographic and clinico-pathological factors on the testing uptake; and to estimate the prevalence of EGFR mutation if all patients were tested. METHODS: This population-based study involved all patients who were diagnosed with non-squamous NSCLC in four health regions in New Zealand between January 2010 and December 2015. Eligible patients were identified from the New Zealand Cancer Registry and information on EGFR mutation testing was obtained through linkage to TestSafe, a clinical information sharing service, and laboratory records. RESULTS: Of 2701 eligible patients, 1059 (39.2%) were tested for EGFR mutation. The testing prevalence increased (3.7% in 2010 to 64.6% in 2014) and the influences of demographic and clinic-pathological factors decreased from 2010 to June 2014, and remained stable afterward. Of the tested patients, 229 (21.6%) were mutation positive with a decreasing trend observed from 2010 (43.8%) to June 2014 (16.8%). The best-fit log-linear model estimated the prevalence of EGFR mutation, if all patients were tested, as 15.5% (95% CI: 13.2%-18.0%). CONCLUSION: The methods described here allowed a more accurate estimation of the prevalence of EGFR mutation.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Pruebas Genéticas/estadística & datos numéricos , Neoplasias Pulmonares/genética , Adulto , Anciano , Receptores ErbB/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Nueva Zelanda , Prevalencia , Sistema de Registros
4.
Pharmaceutics ; 10(3)2018 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-30096910

RESUMEN

Multidrug resistance (MDR) is a major hurdle which must be overcome to effectively treat cancer. ATP-binding cassette transporters (ABC transporters) play pivotal roles in drug absorption and disposition, and overexpression of ABC transporters has been shown to attenuate cellular/tissue drug accumulation and thus increase MDR across a variety of cancers. Overcoming MDR is one desired approach to improving the survival rate of patients. To date, a number of modulators have been identified which block the function and/or decrease the expression of ABC transporters, thereby restoring the efficacy of a range of anticancer drugs. However, clinical MDR reversal agents have thus far proven ineffective and/or toxic. The need for new, effective, well-tolerated and nontoxic compounds has led to the development of natural compounds and their derivatives to ameliorate MDR. This review evaluates whether synthetically modifying natural compounds is a viable strategy to generate potent, nontoxic, ABC transporter inhibitors which may potentially reverse MDR.

5.
Target Oncol ; 13(1): 89-98, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29188408

RESUMEN

BACKGROUND: Delta-like ligand 4-Notch (DLL4-Notch) signaling contributes to the maintenance of chemotherapy-resistant cancer stem cells and tumor vasculature. OBJECTIVE: This phase IB trial of demcizumab, an IgG2 humanized monoclonal antibody directed against DLL4, was undertaken to determine its maximum tolerated dose, safety, immunogenicity, preliminary efficacy, pharmacokinetics, and pharmacodynamics, combined with standard chemotherapy. PATIENTS AND METHODS: Forty-six treatment-naive patients with metastatic non-squamous non-small cell lung cancer (NSCLC) were enrolled in this open-label, dose-escalation study using a standard 6 + 6 design. Demcizumab (2.5, 5.0, and 7.5 mg/kg) was given once every 3 weeks with standard doses of pemetrexed and carboplatin using a continuous (six cycles followed by demcizumab maintenance) or a truncated demcizumab regimen (four cycles followed by pemetrexed maintenance). RESULTS: Initially, continuous demcizumab was given until progression but two patients developed grade 3 pulmonary hypertension and congestive heart failure after eight or more infusions. Thereafter, 23 patients were treated with a truncated regimen of demcizumab, which was not associated with any grade 3 or greater cardiopulmonary toxicity. Common adverse events were hypertension, raised brain natriuretic peptide, and those expected from carboplatin and pemetrexed alone. Twenty of 40 evaluable patients (50%) had objective tumor responses. In peripheral blood, demcizumab treatment modulated the expression of genes regulating Notch signaling and angiogenesis, and achieved concentrations exceeding those saturating DLL4 binding. CONCLUSIONS: This study has identified a truncated dosing regimen and recommended phase II dose of demcizumab (5 mg/kg q3-weekly ×4) for subsequent clinical evaluation in combination with standard carboplatin and pemetrexed chemotherapy. NCT01189968.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Carboplatino/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Pemetrexed/uso terapéutico , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/farmacología , Carboplatino/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Femenino , Humanos , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Pemetrexed/farmacología
6.
Oncotarget ; 8(60): 101437-101451, 2017 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-29254176

RESUMEN

To investigate the clinical validity and utility of tests for detecting Epidermal Growth Factor Receptor (EGFR) gene mutations in non-squamous non-small cell lung cancer patients, tumour DNA extracts from 532 patients previously tested by the cobas EGFR Mutation Test (RT-PCR test) were retested by the Sequenom/Agena Biosciences MassArray OncoFocus mass spectrometry test (MS test). Valid results from both tests were available from 470 patients (88%) for agreement analysis. Survival data were obtained for 513 patients (96%) and 77 patients (14%) were treated with EGFR tyrosine kinase inhibitors (TKIs). Agreement analysis revealed moderately high positive (79.8%), negative (96.9%) and overall percentage agreement (93.2%) for the detection of EGFR mutations. However, EGFR mutations were detected by one test and not by the other test in 32 patients (7%). Retesting of discordant samples revealed false-positive and false-negative results generated by both tests. Despite this, treatment and survival outcomes correlated with the results of the RT-PCR and MS tests. In conclusion, this study provides evidence of the clinical validity and utility of the RT-PCR and MS tests for detection of EGFR mutations that predict prognosis and benefit from EGFR-TKI treatment. However, their false-positive and false-negative test results may have important clinical consequences.

7.
Sci Rep ; 7(1): 4073, 2017 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-28642473

RESUMEN

This study evaluated the impact of calcium and magnesium on the in vitro degradation and in vivo clearance of oxaliplatin. Intact oxaliplatin and Pt(DACH)Cl2 were measured in incubation solutions by HPLC-UV. A clinical study determined changes in plasma concentrations of calcium and magnesium in cancer patients and their impact on oxaliplatin clearance. Kinetic analyses modelled oxaliplatin degradation reactions in vitro and contributions to oxaliplatin clearance in vivo. Calcium and magnesium accelerated oxaliplatin degradation to Pt(DACH)Cl2 in chloride-containing solutions in vitro. Kinetic models based on calcium and magnesium binding to a monochloro-monooxalato ring-opened anionic oxaliplatin intermediate fitted the in vitro degradation time-course data. In cancer patients, calcium and magnesium plasma concentrations varied and were increased by giving calcium gluconate and magnesium sulfate infusions, but did not alter or correlate with oxaliplatin clearance. The intrinsic in vitro clearance of oxaliplatin attributed to chloride-, calcium- and magnesium-mediated degradation predicted contributions of <2.5% to the total in vivo clearance of oxaliplatin. In conclusion, calcium and magnesium accelerate the in vitro degradation of oxaliplatin by binding to a monochloro-monooxalato ring-opened anionic intermediate. Kinetic analysis of in vitro oxaliplatin stability data can be used for in vitro prediction of potential effects on oxaliplatin clearance in vivo.


Asunto(s)
Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Oxaliplatino/metabolismo , Oxaliplatino/farmacocinética , Antineoplásicos/química , Calcio/sangre , Calcio/metabolismo , Cloruros/metabolismo , Humanos , Cinética , Magnesio/sangre , Magnesio/metabolismo , Oxaliplatino/química
8.
J Inorg Biochem ; 177: 249-258, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28551160

RESUMEN

Oxaliplatin-based chemotherapy is the mainstay for the treatment of advanced colorectal cancer. Copper transporter proteins have been implicated in the transport of platinum-based anticancer drugs, but their expression in human colorectal cancer cell lines and roles in controlling their sensitivity to oxaliplatin are not well studied or understood. The endogenous and modified expression of copper uptake transporter 1 (hCTR1) was studied in a panel of human colorectal cancer cell lines (DLD-1, SW620, HCT-15 and COLO205) with ~20-fold variation in oxaliplatin sensitivity. hCTR1 protein was expressed more abundantly than ATP7A and ATP7B proteins, but with broadly similar levels and patterns of expression across four colorectal cancer cell lines. In a colorectal cancer cell-line background (DLD-1), stable transfection of the hCtr1 gene enhanced hCTR1 protein expression and increased the sensitivity of the cells to the cytotoxicity of copper and oxaliplatin. Treatment with copper chelators (ammonium tetrathiomolybdate, bathocuproinedisulfonic acid and D-penicillamine) increased expression of hCTR1 protein in DLD-1 and SW620 cells, and potentiated the cytotoxicity of oxaliplatin in DLD-1 but not SW620 cells. Treatment with copper chloride altered neither the expression of copper transporters nor cytotoxicity of oxaliplatin in colorectal cancer lines. In conclusion, human colorectal cancer cell lines consistently express hCTR1 protein despite their variable sensitivity to oxaliplatin. Genetic or pharmacological modification of hCTR1 protein expression may potentiate oxaliplatin sensitivity in some but not all colorectal cancer cell lines.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Transporte de Catión/genética , Compuestos Organoplatinos/farmacología , Proteínas de Transporte de Catión/metabolismo , Línea Celular Tumoral , Quelantes/farmacología , Cobre/metabolismo , Transportador de Cobre 1 , ATPasas Transportadoras de Cobre/metabolismo , Sinergismo Farmacológico , Humanos , Molibdeno/farmacología , Compuestos Organoplatinos/metabolismo , Oxaliplatino , Penicilamina/farmacología , Fenantrolinas/farmacología , Regulación hacia Arriba/efectos de los fármacos
9.
Expert Opin Drug Metab Toxicol ; 12(12): 1479-1490, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27539508

RESUMEN

INTRODUCTION: Oxaliplatin-based chemotherapy has become the standard treatment for colorectal cancer and other gastrointestinal tumor types. Oxaliplatin-induced neurotoxicity is a major treatment-limiting side effect that compromizes the delivery of cancer treatment and causes long-standing neurological deficits that negatively impact upon patient quality of life Areas covered: The prevention of oxaliplatin-induced neurotoxicity represents an important opportunity for new therapeutic product development to address this major unmet medical need. In this article, we describe a phase Ib clinical trial design, and study procedures and protocols, that we have developed and now propose for the early clinical evaluation of investigational therapeutics for preventing oxaliplatin-induced neurotoxicity. Expert opinion: Recently, several advances have been made in the development of research methodologies applicable to the clinical evaluation of investigational drugs for preventing oxaliplatin-induced neurotoxicity. As we gain better understanding of the mechanisms of oxaliplatin-induced neurotoxicity, we will be able to use these methods to develop and test more effective and targeted neuroprotective agents that may not only improve patients' quality of life but also improve treatment delivery and survival outcomes.


Asunto(s)
Síndromes de Neurotoxicidad/prevención & control , Compuestos Organoplatinos/efectos adversos , Proyectos de Investigación , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Ensayos Clínicos Fase I como Asunto/métodos , Estudios Cruzados , Método Doble Ciego , Neoplasias Gastrointestinales/tratamiento farmacológico , Humanos , Síndromes de Neurotoxicidad/etiología , Compuestos Organoplatinos/administración & dosificación , Oxaliplatino , Calidad de Vida , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos
10.
J Struct Biol ; 192(3): 539-544, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26522274

RESUMEN

The discovery of genetic drivers of lung cancer in patient sub-groups has led to their use as predictive biomarkers and as targets for selective drug therapy. Some of the most important lung cancer drivers are mutations in the EGFR gene, for example, the exon 19 deletions and the L858R variant that confer sensitivity to the front line drugs erlotinib and gefitinib; the acquired T790M variants confer drug resistance and a poor prognosis. A challenge then in targeting EGFR is to produce drugs that inhibit both sensitising variants and resistance variants, leaving wild type protein in healthy cells unaffected. One such agent is AstraZeneca's "breakthrough" AZD9291 molecule that shows a 200-fold selectivity for T790M/L858R over wild type EGFR. Our X-ray crystal structure reveals the binding mode of AZD9291 to the kinase domain of wild type EGFR.


Asunto(s)
Acrilamidas/farmacología , Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Inhibidores de Proteínas Quinasas/farmacología , Acrilamidas/metabolismo , Compuestos de Anilina/metabolismo , Cristalografía por Rayos X , Resistencia a Antineoplásicos/genética , Receptores ErbB/ultraestructura , Clorhidrato de Erlotinib/farmacología , Gefitinib , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Unión Proteica/fisiología , Quinazolinas/farmacología
11.
J Neurochem ; 135(6): 1099-112, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26364854

RESUMEN

Platinum-based anticancer drugs cause peripheral neurotoxicity by damaging sensory neurons within the dorsal root ganglia (DRG), but the mechanisms are incompletely understood. The roles of platinum DNA binding, transcription inhibition and altered cell size were investigated in primary cultures of rat DRG cells. Click chemistry quantitative fluorescence imaging of RNA-incorporated 5-ethynyluridine showed high, but wide ranging, global levels of transcription in individual neurons that correlated with their cell body size. Treatment with platinum drugs reduced neuronal transcription and cell body size to an extent that corresponded to the amount of preceding platinum DNA binding, but without any loss of neuronal cells. The effects of platinum drugs on neuronal transcription and cell body size were inhibited by blocking platinum DNA binding with sodium thiosulfate, and mimicked by treatment with a model transcriptional inhibitor, actinomycin D. In vivo oxaliplatin treatment depleted the total RNA content of DRG tissue concurrently with altering DRG neuronal size. These findings point to a mechanism of chemotherapy-induced peripheral neurotoxicity, whereby platinum DNA damage induces global transcriptional arrest leading in turn to neuronal atrophy. DRG neurons may be particularly vulnerable to this mechanism of toxicity because of their requirements for high basal levels of global transcriptional activity. Findings point to a new stepwise mechanism of chemotherapy-induced peripheral neurotoxicity, whereby platinum DNA damage induces global transcriptional arrest leading in turn to neuronal atrophy. Dorsal root ganglion neurons may be particularly vulnerable to this neurotoxicity because of their high global transcriptional outputs, demonstrated in this study by click chemistry quantitative fluorescence imaging.


Asunto(s)
Daño del ADN/efectos de los fármacos , ADN/efectos de los fármacos , Quimioterapia , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad/tratamiento farmacológico , Compuestos Organoplatinos/farmacología , Platino (Metal)/farmacología , Animales , Proteínas de Transporte de Catión/efectos de los fármacos , Proteínas de Transporte de Catión/metabolismo , Células Cultivadas , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Neuronas/metabolismo , Síndromes de Neurotoxicidad/metabolismo , Oxaliplatino , Ratas
12.
Front Oncol ; 4: 221, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25232533

RESUMEN

The mTOR pathway is a key regulator of multiple cellular signaling pathways and is a potential target for therapy. We have previously developed two hormone-resistant sub-lines of the MCF-7 human breast cancer line, designated TamC3 and TamR3, which were characterized by reduced mTOR signaling, reduced cell volume, and resistance to mTOR inhibition. Here, we show that these lines exhibit increased sensitivity to carboplatin, oxaliplatin, 5-fluorouracil, camptothecin, doxorubicin, paclitaxel, docetaxel, and hydrogen peroxide. The mechanisms underlying these changes have not yet been characterized but may include a shift from glycolysis to mitochondrial respiration. If this phenotype is found in clinical hormone-resistant breast cancers, conventional cytotoxic therapy may be a preferred option for treatment.

13.
Biopharm Drug Dispos ; 35(8): 450-62, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24889085

RESUMEN

The literature concerning the roles of metal transporting solute carriers in the development and progression of human cancer, and in the delivery of metal-containing anticancer drugs, chemical carcinogens and imaging agents, is reviewed. A range of different solute carrier families, including members from the SLC2A, SLC11A, SLC22A, SLC25A, SLC30A, SLC31A, SLC39A, SLC40A, SLC47A and SLCO1B families, and various metal substrates, including arsenic, copper, gadolinium, iron, platinum and zinc, have been implicated in these cancer-related transport processes. For example, the transport of platinum-based anticancer drugs has been reported to be influenced by the expression and activities of OCT1-3 (SLC22A1-3), OCTN1/2 (SLC22A4/5), CTR1/2 (SLC31A1/2) and MATE1/2 (SLC47A1/2) solute carriers. As another example, solute carriers mediate control over the availability of endogenous metal ions, such as copper, iron and zinc, may have key roles in regulating tumour angiogenesis, cell proliferation, epithelial-to-mesenchymal transition and aberrant MAPK and STAT-3 signal transduction in cancer. In conclusion, emerging mechanisms involving metal transporting solute carriers are being defined and seem likely to make major contributions to cancer development and progression, and to the delivery of anticancer and tumour imaging agents.


Asunto(s)
Proteínas de Transporte de Anión/metabolismo , Antineoplásicos/metabolismo , Proteínas de Transporte de Catión/metabolismo , Metales/metabolismo , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Transporte Biológico , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Humanos , Translocasas Mitocondriales de ADP y ATP/metabolismo , Neoplasias/tratamiento farmacológico , Distribución Tisular
14.
Cancer Chemother Pharmacol ; 74(1): 25-35, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24801172

RESUMEN

PURPOSE: SN 28049 is a new DNA-binding topoisomerase II poison identified by its curative activity against the murine colon 38 carcinoma. Previous studies showed activity to be associated with selective drug accumulation and retention in tumour tissue. Retention varied widely among different tumours and was related to antitumour activity. We determined whether differences in the uptake and retention of SN 28049 could be observed in vitro. METHODS: The Co38P and LLTC lines were derived from the murine colon 38 carcinoma and Lewis lung carcinoma (3LL), respectively. The NZM4, NZM10 and NZM52 human melanoma lines, as well as the CCRF/CEM, CEM/VLB100 and CEM/E1000 human leukaemia lines were also utilised. Cell-associated drug was measured by liquid chromatography-mass spectrometry, laser-scanning confocal microscopy and fluorescence microscopy. Data for SN 28049 were compared for four SN 28049 analogues, for the structurally related drug N-[2-(dimethylamino)-ethyl]acridine-4-carboxamide (DACA) and for doxorubicin. RESULTS: Cellular uptake of SN 28049 was rapid and associated with increased fluorescence in cytoplasmic vesicles or bodies. SN 28049 uptake after an incubation time of 1 h varied widely with different cell lines (2-98 pmol/106 cells) and did not correlate with growth inhibitory concentrations (IC50 values), which also varied widely (1.2-19 nM). Changes in the length of the N-linked side chain of SN 28049 had large effects on drug uptake by Co38P cells. SN 28049 uptake by CCRF/CEM cells was only slightly affected by the expression of P-glycoprotein (CEM/VLB100) or MRP1 protein (CEM/E1000). As measured by cytoplasmic fluorescence, SN 28049 was taken up rapidly and retained strongly by Co38P cells, DACA was taken up rapidly and retained poorly, and doxorubicin was taken up slowly and retained moderately. CONCLUSIONS: The results suggest that SN 28049 is actively transported into cytoplasmic vesicles. While vesicle-associated drug is not important for intrinsic cytotoxicity, it may play a key role as a "slow release" form that modifies pharmacokinetics in multicellular structures such as tumours.


Asunto(s)
Antineoplásicos/metabolismo , Carcinoma/metabolismo , Drogas en Investigación/metabolismo , Leucemia/metabolismo , Melanoma/metabolismo , Naftiridinas/metabolismo , Inhibidores de Topoisomerasa II/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Transporte Biológico Activo , Carcinoma/tratamiento farmacológico , Carcinoma/ultraestructura , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Vesículas Citoplasmáticas/efectos de los fármacos , Vesículas Citoplasmáticas/metabolismo , Vesículas Citoplasmáticas/ultraestructura , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Drogas en Investigación/química , Drogas en Investigación/farmacología , Humanos , Cinética , Leucemia/tratamiento farmacológico , Leucemia/patología , Melanoma/tratamiento farmacológico , Melanoma/ultraestructura , Ratones , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Naftiridinas/química , Naftiridinas/farmacología , Proteínas de Neoplasias/metabolismo , Inhibidores de Topoisomerasa II/química , Inhibidores de Topoisomerasa II/farmacología
15.
BMC Cancer ; 13: 495, 2013 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-24156389

RESUMEN

BACKGROUND: Calcium and magnesium (Ca/Mg) infusions have been suggested as an effective intervention for preventing oxaliplatin-induced neurotoxicity, but the effects of Ca/Mg infusions on oxaliplatin pharmacokinetics, motor nerve hyperexcitability and acute neurotoxicity symptoms are unclear. METHODS: In this double blind crossover study, colorectal cancer patients undergoing oxaliplatin-based chemotherapy were randomised to receive Ca/Mg (1g Ca Gluconate plus 1g MgSO4) on cycle 1 and placebo (vehicle alone) on cycle 2, or to receive the same treatments in the opposite sequence. Study endpoints included plasma pharmacokinetics of intact oxaliplatin and free platinum; electromyography (EMG) detection of abnormal spontaneous high-frequency motor unit action potential discharges; and patient-reported acute neurotoxicity symptoms and their preferred study treatment for reducing these symptoms. RESULTS: Nineteen of 20 enrolled patients completed the study. Plasma pharmacokinetics of intact oxaliplatin and free platinum were similar when oxaliplatin was given with Ca/Mg or placebo (ratio of geometric means of AUC0-t with Ca/Mg or placebo: intact oxaliplatin, 0.95 (90% CI, 0.90 - 1.01); free platinum, 0.99 (90% CI, 0.94 - 1.05)). EMG motor nerve hyperexcitability scores were similar with Ca/Mg and placebo (mean difference in EMG score between Ca/Mg and placebo: -0.3 (95% CI, -2.2 - 1.6)). Patient-reported acute neurotoxicity symptoms were similar in frequency with Ca/Mg and placebo. For reducing neurotoxic symptoms, fewer patients preferred Ca/Mg than placebo or neither treatment (26% versus 74%; P<0.01). CONCLUSIONS: Ca/Mg infusions do not alter the clinical pharmacokinetics of oxaliplatin and do not seem to reduce its acute neurotoxicity. TRIAL REGISTRATION: Trial registration identifier ACTRN12611000738921.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Calcio/administración & dosificación , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/tratamiento farmacológico , Magnesio/administración & dosificación , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Adulto , Anciano , Neoplasias Colorrectales/patología , Estudios Cruzados , Interacciones Farmacológicas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Compuestos Organoplatinos/administración & dosificación , Compuestos Organoplatinos/farmacocinética , Compuestos Organoplatinos/toxicidad , Oxaliplatino , Enfermedades del Sistema Nervioso Periférico/diagnóstico , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Resultado del Tratamiento
16.
Clin Exp Pharmacol Physiol ; 40(6): 371-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23556474

RESUMEN

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


Asunto(s)
Proteínas de Transporte de Catión/antagonistas & inhibidores , Ganglios Espinales/efectos de los fármacos , Intoxicación del Sistema Nervioso por Metales Pesados/prevención & control , Histidina/análogos & derivados , Compuestos Organometálicos/farmacología , Compuestos Organoplatinos/toxicidad , Platino (Metal)/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Transportador de Cobre 1 , ATPasas Transportadoras de Cobre , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Femenino , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Células HEK293 , Intoxicación del Sistema Nervioso por Metales Pesados/patología , Histidina/farmacología , Humanos , Compuestos Organoplatinos/farmacocinética , Oxaliplatino , Ratas , Transfección
17.
Biochem Pharmacol ; 85(2): 207-15, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23123662

RESUMEN

Dorsal root ganglion (DRG) neurons are affected by platinum-induced neurotoxicity and neurodegenerative processes associated with disturbed copper homeostasis and transport. This study aimed to understand the role of copper transporter 1 (Ctr1) in the uptake and toxicity of copper and platinum drugs in cultured rat DRG neurons, and the functional activities of rat Ctr1 (rCtr1) as a membrane transporter of copper and platinum drugs. Heterologous expression of rCtr1 in HEK293 cells (HEK/rCtr1 cells) increased the uptake and cytotoxicity of copper, oxaliplatin, cisplatin and carboplatin, in comparison to isogenic vector-transfected control cells. Cultured rat DRG neurons endogenously expressed rCtr1 protein on their neuronal cell body plasma membranes and cytoplasm, and displayed substantial capacity for taking up copper, but were resistant to copper toxicity. The uptake of copper by both cultured rat DRG neurons and HEK/rCtr1 cells was saturable and inhibited by cold temperature, silver and zinc, consistent with it being mediated by rCtr1. Cultured rat DRG neurons accumulated platinum during their exposure to oxaliplatin and were sensitive to oxaliplatin cytotoxicity. The accumulation of platinum by both cultured rat DRG neurons and HEK/rCtr1 cells, during oxaliplatin exposure, was saturable and temperature dependent, but was inhibited by copper only in HEK/rCtr1 cells. In conclusion, rCtr1 can transport copper and platinum drugs, and sensitizes cells to their cytotoxicities. DRG neurons display substantial capacity for accumulating copper via a transport process mediated by rCtr1, but appear able to resist copper toxicity and use alternative mechanisms to take up oxaliplatin.


Asunto(s)
Antineoplásicos/metabolismo , Proteínas de Transporte de Catión/metabolismo , Cobre/efectos adversos , Ganglios Espinales/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Compuestos Organoplatinos/metabolismo , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Transporte Biológico/efectos de los fármacos , Carboplatino/efectos adversos , Carboplatino/metabolismo , Carboplatino/farmacología , Proteínas de Transporte de Catión/antagonistas & inhibidores , Proteínas de Transporte de Catión/genética , Células Cultivadas , Cisplatino/efectos adversos , Cisplatino/metabolismo , Cisplatino/farmacología , Cobre/metabolismo , Cobre/farmacología , Sulfato de Cobre/efectos adversos , Sulfato de Cobre/metabolismo , Sulfato de Cobre/farmacología , Transportador de Cobre 1 , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Células HEK293 , Humanos , Concentración 50 Inhibidora , Moduladores del Transporte de Membrana/farmacología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Neuronas/metabolismo , Compuestos Organoplatinos/efectos adversos , Compuestos Organoplatinos/farmacología , Oxaliplatino , Ratas , Ratas Wistar , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo
18.
BMC Cancer ; 12: 496, 2012 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-23098625

RESUMEN

BACKGROUND: The purpose of this phase Ib clinical trial was to determine the maximum tolerated dose (MTD) of PR-104 a bioreductive pre-prodrug given in combination with gemcitabine or docetaxel in patients with advanced solid tumours. METHODS: PR-104 was administered as a one-hour intravenous infusion combined with docetaxel 60 to 75 mg/m2 on day one given with or without granulocyte colony stimulating factor (G-CSF) on day two or administrated with gemcitabine 800 mg/m2 on days one and eight, of a 21-day treatment cycle. Patients were assigned to one of ten PR-104 dose-levels ranging from 140 to 1100 mg/m2 and to one of four combination groups. Pharmacokinetic studies were scheduled for cycle one day one and 18F fluoromisonidazole (FMISO) positron emission tomography hypoxia imaging at baseline and after two treatment cycles. RESULTS: Forty two patients (23 females and 19 males) were enrolled with ages ranging from 27 to 85 years and a wide range of advanced solid tumours. The MTD of PR-104 was 140 mg/m2 when combined with gemcitabine, 200 mg/m2 when combined with docetaxel 60 mg/m2, 770 mg/m2 when combined with docetaxel 60 mg/m2 plus G-CSF and ≥770 mg/m2 when combined with docetaxel 75 mg/m2 plus G-CSF. Dose-limiting toxicity (DLT) across all four combination settings included thrombocytopenia, neutropenic fever and fatigue. Other common grade three or four toxicities included neutropenia, anaemia and leukopenia. Four patients had partial tumour response. Eleven of 17 patients undergoing FMISO scans showed tumour hypoxia at baseline. Plasma pharmacokinetics of PR-104, its metabolites (alcohol PR-104A, glucuronide PR-104G, hydroxylamine PR-104H, amine PR-104M and semi-mustard PR-104S1), docetaxel and gemcitabine were similar to that of their single agents. CONCLUSIONS: Combination of PR-104 with docetaxel or gemcitabine caused dose-limiting and severe myelotoxicity, but prophylactic G-CSF allowed PR-104 dose escalation with docetaxel. Dose-limiting thrombocytopenia prohibited further evaluation of the PR104-gemcitabine combination. A recommended dose was identified for phase II trials of PR-104 of 770 mg/m2 combined with docetaxel 60 to 75 mg/m2 both given on day one of a 21-day treatment cycle supported by prophylactic G-CSF (NCT00459836).


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias/tratamiento farmacológico , Profármacos/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Área Bajo la Curva , Hipoxia de la Célula/fisiología , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Docetaxel , Relación Dosis-Respuesta a Droga , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Femenino , Semivida , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/sangre , Neoplasias/metabolismo , Compuestos de Mostaza Nitrogenada/administración & dosificación , Compuestos de Mostaza Nitrogenada/efectos adversos , Compuestos de Mostaza Nitrogenada/sangre , Compuestos de Mostaza Nitrogenada/farmacocinética , Profármacos/efectos adversos , Taxoides/administración & dosificación , Taxoides/farmacocinética , Gemcitabina
19.
Curr Cancer Drug Targets ; 12(8): 962-86, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22794121

RESUMEN

Membrane transporters govern the movement of drugs and their metabolites across biological membranes, thereby determining their pharmacokinetics, efficacy and adverse drug reactions. Platinum-based anticancer drugs are a mainstay of chemotherapy for many human malignancies. However, their clinical utility is limited by tumor resistance and normal tissue toxicities, which are determined at least in part by the level of tissue accumulation of platinum. Recently, several members of the ATP-binding cassette (ABC), solute carrier (SLC) and ATPase membrane protein superfamilies have been found to contribute to the net accumulation of platinum drugs in malignant and normal tissues. Herein, a review has been carried out to critically evaluate current preclinical and clinical evidence implicating membrane transporters as determinants of the pharmacology of cisplatin, oxaliplatin, carboplatin and related investigational compounds. The evidence includes studies of recombinant cell systems with genetically modified expression of individual membrane transporters, platinum-resistant or -sensitive human cancer cells and in vivo xenografted tumors, animal models of platinum-induced nephro-, oto- or neurotoxicity, and clinical studies of associations between the membrane transporter tumor expression and patient outcomes from platinum-based chemotherapy. Understanding the role of membrane transporters as determinants of the pharmacology of platinum drugs will be a basis for targeting these drug transporters in individualized and optimized platinum-based cancer therapy, and new drug development.


Asunto(s)
Antineoplásicos/farmacocinética , Proteínas de Transporte de Membrana/metabolismo , Compuestos de Platino/farmacología , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Animales , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Cisplatino/farmacología , Cisplatino/toxicidad , ATPasas Transportadoras de Cobre , Humanos , Proteínas de Transporte de Membrana/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Platino (Metal)/farmacología , Platino (Metal)/uso terapéutico , Platino (Metal)/toxicidad , Compuestos de Platino/farmacocinética , Proteínas SLC31 , Resultado del Tratamiento
20.
BMC Cancer ; 11: 432, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21982454

RESUMEN

BACKGROUND: The phosphate ester PR-104 is rapidly converted in vivo to the alcohol PR-104A, a nitrogen mustard prodrug that is metabolised to hydroxylamine (PR-104H) and amine (PR-104M) DNA crosslinking agents by one-electron reductases in hypoxic cells and by aldo-keto reductase 1C3 independently of oxygen. In a previous phase I study using a q 3 week schedule of PR-104, the maximum tolerated dose (MTD) was 1100 mg/m2 and fatigue, neutropenic fever and infection were dose-limiting. The primary objective of the current study was to determine the dose-limiting toxicity (DLT) and MTD of weekly PR-104. METHODS: Patients with advanced solid tumours received PR-104 as a 1-hour intravenous infusion on days 1, 8 and 15 every 28 days with assessment of pharmacokinetics on cycle 1 day 1. Twenty-six patients (pts) were enrolled (16 male/10 female; median age 58 yrs, range 30 to 70 yrs) who had received a median of two prior chemotherapy regimens (range, 0 to 3) for melanoma (8 pts), colorectal or anal cancer (3 pts), NSCLC (3 pts), sarcoma (3 pts), glioblastoma (2 pts), salivary gland tumours (2 pts) or other solid tumours (5 pts). PR-104 was administered at 135 mg/m2 (3 pts), 270 mg/m2 (6 pts), 540 mg/m2 (6 pts), 675 mg/m2 (7 pts) and 900 mg/m2 (4 pts) for a median of two treatment cycles (range, 1 to 7 cycles) and five infusions (range, 1 to 18) per patient. RESULTS: Dose-limiting toxicities (DLTs) during cycle one included grade four thrombocytopenia at 540 mg/m2 (1 of 6 pts) and grade four thrombocytopenia and neutropenia at 900 mg/m2 (2 of 4 pts). At an intermediate dose of 675 mg/m2, there were no DLTs among a total of seven patients given 12 treatment cycles but all experienced moderate to severe (grade 2 to 4) haematological toxicity. Thrombocytopenia was delayed in its onset and nadir, and its recovery was protracted and incomplete in many patients. There were no complete or partial tumour responses. PR-104-induced thrombocytopenia and neutropenia correlated with plasma AUC of PR-104, PR-104A and an oxidative semi-mustard metabolite (PR-104S1), but no more strongly than with PR-104 dose-level. There was no significant correlation between plasma AUC for the reduced metabolites and myelotoxicity. CONCLUSIONS: Thrombocytopenia, and to a lesser extent neutropenia, was the DLT of weekly PR-104. The MTD was 675 mg/m2/week. PR-104 given weekly may be a suitable protocol for further clinical evaluation as a short course of treatment with fractionated radiotherapy or haematopoietic stem cell support, as its duration of dosing is restricted by delayed-onset and protracted thrombocytopenia.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Compuestos de Mostaza Nitrogenada/uso terapéutico , Profármacos/uso terapéutico , Adulto , Anciano , Antineoplásicos/farmacología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Compuestos de Mostaza Nitrogenada/farmacología , Profármacos/farmacología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA