Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Br J Cancer ; 122(12): 1760-1768, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32350413

RESUMEN

BACKGROUND: Nab-paclitaxel plus gemcitabine (nabP+gemcitabine) offers modest survival gains for patients with metastatic pancreatic ductal adenocarcinoma (PDAC). Sequential scheduling of nabP+gemcitabine in a PDAC mouse model improved efficacy; this hypothesis was tested in a clinical trial. METHODS: Patients with previously untreated metastatic PDAC were randomised to receive nabP+gemcitabine administered either concomitantly on the same day, or sequentially, with gemcitabine administered 24 h after nabP. The primary outcome measure was progression-free survival (PFS). Secondary outcome measures were objective response rate (ORR), overall survival (OS), safety, quality of life (QoL) and predictive biomarkers. RESULTS: In total, 71 patients received sequential (SEQ) and 75 concomitant (CON) treatment. Six-month PFS was 46% with SEQ and 32% with CON scheduling. Median PFS (5.6 versus 4.0 months, hazard ratio [HR] 0.67, 95% confidence interval [95% CI] 0.47-0.95, p = 0.022) and ORR (52% versus 31%, p = 0.023) favoured the SEQ arm; median OS was 10.2 versus 8.2 months (HR 0.93, 95% CI 0.65-1.33, p = 0.70). CTCAE Grade ≥3 neutropaenia incidence doubled with SEQ therapy but was not detrimental to QoL. Strongly positive tumour epithelial cytidine deaminase (CDA) expression favoured benefit from SEQ therapy (PFS HR 0.31, 95% CI 0.13-0.70). CONCLUSIONS: SEQ delivery of nabP+gemcitabine improved PFS and ORR, with manageable toxicity, but did not significantly improve OS. CLINICAL TRIAL REGISTRATION: ISRCTN71070888; ClinialTrials.gov (NCT03529175).


Asunto(s)
Albúminas/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Carcinoma Ductal Pancreático/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Paclitaxel/administración & dosificación , Neoplasias Pancreáticas/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/mortalidad , Desoxicitidina/administración & dosificación , Esquema de Medicación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/mortalidad , Supervivencia sin Progresión , Gemcitabina , Neoplasias Pancreáticas
2.
Gut ; 67(3): 497-507, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28077438

RESUMEN

OBJECTIVE: Desmoplasia and hypovascularity are thought to impede drug delivery in pancreatic ductal adenocarcinoma (PDAC). However, stromal depletion approaches have failed to show clinical responses in patients. Here, we aimed to revisit the role of the tumour microenvironment as a physical barrier for gemcitabine delivery. DESIGN: Gemcitabine metabolites were analysed in LSL-KrasG12D/+ ; LSL-Trp53R172H/+ ; Pdx-1-Cre (KPC) murine tumours and matched liver metastases, primary tumour cell lines, cancer-associated fibroblasts (CAFs) and pancreatic stellate cells (PSCs) by liquid chromatography-mass spectrometry/mass spectrometry. Functional and preclinical experiments, as well as expression analysis of stromal markers and gemcitabine metabolism pathways were performed in murine and human specimen to investigate the preclinical implications and the mechanism of gemcitabine accumulation. RESULTS: Gemcitabine accumulation was significantly enhanced in fibroblast-rich tumours compared with liver metastases and normal liver. In vitro, significantly increased concentrations of activated 2',2'-difluorodeoxycytidine-5'-triphosphate (dFdCTP) and greatly reduced amounts of the inactive gemcitabine metabolite 2',2'-difluorodeoxyuridine were detected in PSCs and CAFs. Mechanistically, key metabolic enzymes involved in gemcitabine inactivation such as hydrolytic cytosolic 5'-nucleotidases (Nt5c1A, Nt5c3) were expressed at low levels in CAFs in vitro and in vivo, and recombinant expression of Nt5c1A resulted in decreased intracellular dFdCTP concentrations in vitro. Moreover, gemcitabine treatment in KPC mice reduced the number of liver metastases by >50%. CONCLUSIONS: Our findings suggest that fibroblast drug scavenging may contribute to the clinical failure of gemcitabine in desmoplastic PDAC. Metabolic targeting of CAFs may thus be a promising strategy to enhance the antiproliferative effects of gemcitabine.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacocinética , Carcinoma Ductal Pancreático/metabolismo , Desoxicitidina/análogos & derivados , Fibroblastos/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Pancreáticas/metabolismo , 5'-Nucleotidasa/metabolismo , Actinas/metabolismo , Animales , Antimetabolitos Antineoplásicos/uso terapéutico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/secundario , Línea Celular Tumoral , Citidina Trifosfato/análogos & derivados , Citidina Trifosfato/metabolismo , Desoxicitidina/farmacocinética , Desoxicitidina/uso terapéutico , Floxuridina/análogos & derivados , Floxuridina/metabolismo , Humanos , Hígado/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/secundario , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Cultivo Primario de Células , Microambiente Tumoral , Gemcitabina
4.
Br J Cancer ; 111(2): 318-25, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24874484

RESUMEN

BACKGROUND: The modest benefits of gemcitabine (dFdC) therapy in patients with pancreatic ductal adenocarcinoma (PDAC) are well documented, with drug delivery and metabolic lability cited as important contributing factors. We have used a mouse model of PDAC: KRAS(G12D); p53(R172H); pdx-Cre (KPC) that recapitulates the human disease to study dFdC intra-tumoural metabolism. METHODS: LC-MS/MS and NMR were used to measure drug and physiological analytes. Cytotoxicity was assessed by the Sulphorhodamine B assay. RESULTS: In KPC tumour tissue, we identified a new, Kennedy pathway-linked dFdC metabolite (gemcitabine diphosphate choline (GdPC)) present at equimolar amounts to its precursor, the accepted active metabolite gemcitabine triphosphate (dFdCTP). Utilising additional subcutaneous PDAC tumour models, we demonstrated an inverse correlation between GdPC/dFdCTP ratios and cytidine triphosphate (CTP). In tumour homogenates in vitro, CTP inhibited GdPC formation from dFdCTP, indicating competition between CTP and dFdCTP for CTP:phosphocholine cytidylyltransferase (CCT). As the structure of GdPC precludes entry into cells, potential cytotoxicity was assessed by stimulating CCT activity using linoleate in KPC cells in vitro, leading to increased GdPC concentration and synergistic growth inhibition after dFdC addition. CONCLUSIONS: GdPC is an important element of the intra-tumoural dFdC metabolic pathway in vivo.


Asunto(s)
Carcinoma Ductal Pancreático/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Antimetabolitos Antineoplásicos/farmacocinética , Antimetabolitos Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/metabolismo , Colina/metabolismo , Desoxicitidina/metabolismo , Desoxicitidina/farmacocinética , Desoxicitidina/farmacología , Difosfatos/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Pancreáticas/metabolismo , Células Tumorales Cultivadas , Gemcitabina
5.
Br J Cancer ; 107(10): 1692-701, 2012 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-23037716

RESUMEN

BACKGROUND: Amplification of aurora kinase A (AK-A) overrides the mitotic spindle assembly checkpoint, inducing resistance to taxanes. RNA interference targeting AK-A in human pancreatic cancer cell lines enhanced taxane chemosensitivity. In this study, a novel AK-A inhibitor, CYC3, was investigated in pancreatic cancer cell lines, in combination with paclitaxel. METHODS: Western blot, flow cytometry and immunostaining were used to investigate the specificity of CYC3. Sulforhodamine B staining, time-lapse microscopy and colony-formation assays were employed to evaluate the cytotoxic effect of CYC3 and paclitaxel. Human colony-forming unit of granulocyte and macrophage (CFU-GM) cells were used to compare the effect in tumour and normal tissue. RESULTS: CYC3 was shown to be a specific AK-A inhibitor. Three nanomolar paclitaxel (growth inhibition 50% (GI(50)) 3 nM in PANC-1, 5.1 nM in MIA PaCa-2) in combination with 1 µM CYC3 (GI(50) 1.1 µM in MIA PaCa2 and 2 µM in PANC-1) was synergistic in inhibiting pancreatic cell growth and causing mitotic arrest, achieving similar effects to 10-fold higher concentrations of paclitaxel (30 nM). In CFU-GM cells, the effect of the combination was simply additive, displaying significantly less myelotoxicity compared with high concentrations of paclitaxel (30 nM; 60-70% vs 100% inhibition). CONCLUSION: The combination of lower doses of paclitaxel and CYC3 merits further investigation with the potential for an improved therapeutic index in vivo.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Células de la Médula Ósea/efectos de los fármacos , Paclitaxel/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Aurora Quinasa A , Aurora Quinasas , Células de la Médula Ósea/enzimología , Células de la Médula Ósea/patología , Línea Celular Tumoral , Sinergismo Farmacológico , Células Progenitoras de Granulocitos y Macrófagos/efectos de los fármacos , Células Progenitoras de Granulocitos y Macrófagos/metabolismo , Humanos , Paclitaxel/administración & dosificación , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/patología , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Serina-Treonina Quinasas/metabolismo , Células Madre/efectos de los fármacos , Células Madre/enzimología , Células Madre/patología
6.
Eur J Cancer Care (Engl) ; 19(1): 80-90, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19708939

RESUMEN

The introduction, 30 years ago, of the co-administration of appropriate hydration and ensuring a diuresis occurs during the administration of cisplatin was important in its development, allowing clinically significant doses to be given with acceptable rates of toxicity. The clinical usage of cisplatin has increased and hydration protocols have been amended to increase patient comfort and reduce resource utilization. We suspected that this had led to unnecessary variations in practice both in clinical trials and subsequently in the clinic. Therefore, we reviewed practice in the Edinburgh Cancer Centre and discovered that 25 different hydration protocols were in use, with wide variation in dilution of cisplatin, total fluid administered, use of electrolyte (potassium and magnesium) supplementation and diuretics. These differences are a reflection of adoption of variations in hydration regimes published in pivotal clinical trials. A review of the available evidence relating to cisplatin associated hydration regimens was performed and recommendations will be made for the future design of evidence-based protocols.


Asunto(s)
Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Protocolos Clínicos , Fluidoterapia/métodos , Neoplasias/tratamiento farmacológico , Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Diuréticos/administración & dosificación , Esquema de Medicación , Humanos , Escocia
7.
Ann Oncol ; 19(7): 1340-1346, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18325912

RESUMEN

BACKGROUND: Tumour necrosis factor-alpha (TNF-alpha) is an important regulator of the chronic inflammation contributing to tumour progression. Infliximab, an anti-TNF-alpha monoclonal antibody was investigated in this trial of patients with advanced cancer. The primary objectives were to determine the safety profile and biological response of infliximab in a cancer population. Clinical response was a secondary objective. PATIENTS AND METHODS: Forty-one patients received infliximab at 5 mg/kg (n = 21) or 10 mg/kg (n = 20) i.v. at 0 and 2 weeks and then every 4 weeks. Post-treatment samples were measured for changes in plasma and serum TNF-alpha, CCL2, IL-6 and C-reactive protein (CRP). RESULTS: Infliximab was well tolerated with no dose-limiting toxic effects. At both doses of infliximab, neutralisation of serum TNF-alpha was observed after 1 h while plasma CCL2, IL-6 and serum CRP were decreased 24 and 48 h following infliximab administration. Seven patients experienced disease stablisation (range 10-50+ weeks). There was no evidence of disease acceleration in any patient. CONCLUSIONS: Infliximab treatment was safe and well tolerated in patients with advanced cancer. There was evidence of biological activity with baseline TNF-alpha and CCL2 being correlated with infliximab response.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Hipersensibilidad a las Drogas , Hipersensibilidad Tardía , Neoplasias/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adulto , Anciano , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Proteína C-Reactiva/análisis , Quimiocina CCL2/sangre , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Humanos , Hipersensibilidad Tardía/inducido químicamente , Infliximab , Infusiones Intravenosas , Interleucina-6/sangre , Modelos Lineales , Masculino , Persona de Mediana Edad , Neoplasias/sangre , Neoplasias/patología , Sensibilidad y Especificidad , Estomatitis/inducido químicamente , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/sangre
8.
Pharmacogenomics J ; 8(4): 289-96, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17563765

RESUMEN

ATP-binding cassette transporter P-glycoprotein (ABCB1) is responsible for the multidrug resistance (MDR1) phenotype observed in cancer cells. SJG-136, a new pyrrolobenzodiazepine dimer, is a sequence-dependent DNA crosslinking agent and substrate of ABCB1. We previously showed that colon cancer cell lines expressing high levels of ABCB1 showed a lower sensitivity to SJG-136. Here, we show that in 3T3 isogenic fibroblasts, ABCB1 genetic polymorphism differentially affects ABCB1 gene expression and transport function. However, this genotype-phenotype relationship was not observed in immortalized lymphocytes, which expressed 10- to 1000-fold less ABCB1 than colon cancer cell lines. Consistent with this, the cytotoxicity of SJG-136 in 3T3 fibroblasts was affected by ABCB1 genetic polymorphism but not in immortalized lymphocytes. ABCB1 genetic polymorphism is therefore likely to affect drug sensitivity in tissues expressing high levels of the transporter and in which significant variability is observed.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Benzodiazepinas/farmacología , Benzodiazepinonas/farmacología , Polimorfismo Genético/genética , Pirroles/farmacología , Células 3T3 , Subfamilia B de Transportador de Casetes de Unión a ATP , Animales , Benzodiazepinas/química , Benzodiazepinonas/química , Línea Celular Transformada , Línea Celular Tumoral , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Ratones , Polimorfismo Genético/efectos de los fármacos , Pirroles/química
9.
Cancer Chemother Pharmacol ; 60(5): 651-60, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17273826

RESUMEN

Bcl-xL is an anti-apoptotic protein over-expressed in colorectal cancers acting on both the intrinsic and extrinsic pathways. We stably expressed four different short hairpin RNA (pSNG-xL1-4) targeting Bcl-xL in HCT 116 cells. HCT 116 pSNG-xL#1 produced a modest (30%) decrease in Bcl-xL expression whilst Bcl-2 levels were similar to the parental cell line, HCT 116 pSNG-xL#2 and 3 showed 50% decrease in Bcl-xL and stable Bcl-2. HCT 116 pSNG-xL#3 showed a concomitant decrease (50%) in Bcl-2. A decrease in Bcl-xL sensitised cells to the small molecule inhibitor of Bcl-xL, Antimycin A3 and the DNA topoisomerase I inhibitors, SN-38 and camptothecin, but not to doxorubicin. HCT 116 pSNG-xL#1 produced a moderate increase in both senescence and apoptosis and a limited increase in SN-38 induced cell death while HCT 116 pSNG-xL#2 produced an increase in apoptosis but reduced senescence. Finally, when both Bcl-xL and Bcl-2 were decreased to a similar degree (HCT 116 pSNG-xL#3), senescence was significantly increased but apoptosis was limited. This effect was confirmed in vivo after administration of irinotecan and was associated with greater anti-tumour effect. Optimal growth inhibitory effect was therefore observed when both Bcl-xL and Bcl-2 were decreased to a similar extent. Antimycin A3, in combination with SN-38 recapitulated this phenotype in HCT 116 cells, suggesting a potential role for small molecule inhibitors of Bcl-xL/Bcl-2 in the treatment of colorectal cancer, potentially in combination with irinotecan.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Camptotecina/análogos & derivados , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , ARN Interferente Pequeño/farmacología , Proteína bcl-X/genética , Animales , Antimicina A/análogos & derivados , Antimicina A/farmacología , Apoptosis , Camptotecina/farmacología , Muerte Celular , Regulación hacia Abajo , Doxorrubicina/farmacología , Células HCT116 , Humanos , Irinotecán , Ratones , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína bcl-X/metabolismo
10.
Biochem Pharmacol ; 71(4): 408-15, 2006 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-16360645

RESUMEN

Novel ruthenium(II) organo-metallic compounds are active in ovarian cancer models [Aird RE, Cummings J, Ritchie AA, Muir M, Morris RE, Chen H, et al. In vitro and in vivo activity and cross resistance profiles of novel ruthenium(II) organometallic arene complexes in human ovarian cancer. Br J Cancer 2002;86(10):1652-7]. [(eta6-C6H5C6H5)Ru(en)Cl]+ (as a PF6 salt, where en=ethylenediamine (RM175)) has been evaluated in a 13-cell line panel. Particular sensitivity (approximately 10-fold lower than mean IC50) was noted in breast cancer and non-small cell lung cancer cell lines. In addition, IC50 in the A549 was 2 microM and RM175 (25 mg kg-1, days 1 and 5, i.p.) caused a significant (p=0.004) growth delay in a xenograft model. HC11 [(eta6-tetrahydroanthracene)Ru(en)Cl]PF6 was more potent in the A549 cell line (IC50 0.5 microM). HC11 (25 mg kg-1, days 1, 8 and 15, i.p.) was also active in vivo. Following RM175 25 mg kg-1, days 1 and 5, and 15 mg kg-1, days 1-5, HC11 25 and 40 mg kg-1, day 1, elevated alanine transaminase levels were detected, suggesting hepatotoxicity. No changes were observed in kidney or haematological parameters. In liver sections, multi-focal hepatic necrosis was seen, becoming confluent at high doses of HC11. In vitro studies confirmed that HC11 was more toxic than RM175 to fresh human hepatocytes and equitoxic to mithramycin. Liver toxicity may be related to the arene ligand and modification may reduce the potential for hepatic toxicity, while maintaining the anti-tumour activity seen.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Compuestos Organometálicos/farmacología , Compuestos de Rutenio/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Alanina Transaminasa/sangre , Animales , Antineoplásicos/química , Antineoplásicos/toxicidad , Carcinoma de Células Grandes/tratamiento farmacológico , Carcinoma de Células Grandes/patología , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Inyecciones Intraperitoneales , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/ultraestructura , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Desnudos , Compuestos Organometálicos/química , Compuestos Organometálicos/toxicidad , Compuestos de Rutenio/química , Compuestos de Rutenio/toxicidad , Pérdida de Peso/efectos de los fármacos
11.
Eur J Cancer ; 41(12): 1811-8, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16046116

RESUMEN

SJG-136 is a novel pyrrolobenzodiazepine dimer analogue that acts as a minor-groove interstrand DNA cross-linking agent. The present study investigated the impact of ABCB1 (mdr-1) expression on the activity of SJG-136 using both in vitro and in vivo systems. SJG-136 was highly potent in the colon cancer cell lines HCT-116, HT-29 and SW620 (IC50 0.1-0.3 nM). However, HCT-8 and HCT-15 cells expressing significant levels of mdr-1 were less sensitive (IC50 2.3 and 3.7 nM, respectively) using a SRB assay. The cytotoxicity was increased in HCT-15 and A2780(AD) in presence of 5 microg/ml verapamil. Mdr-1 mRNA expression was determined by qRT-PCR and correlated to SJG-136 IC50s (r2=0.86, P=0.0001). Isogenic 3T3 cells expressing mdr-1 cDNA (3T3 pHamdr-1) were less sensitive to SJG-136 than the parental 3T3 cells (IC50 208 and 6.3 nM, respectively). Finally, SJG-136 (120 microg/kg/d dx5) was highly active against A2780 xenografts (SGD=275) but not A2780(AD) xenografts (SGD=67).


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Antineoplásicos/uso terapéutico , Benzodiazepinonas/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , Pirroles/uso terapéutico , Animales , Antineoplásicos/metabolismo , Benzodiazepinonas/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Combinación de Medicamentos , Humanos , Immunoblotting , Concentración 50 Inhibidora , Ratones , Ratones Endogámicos C57BL , Pirroles/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Trasplante Heterólogo , Verapamilo/farmacología
12.
Eur J Cancer ; 41(11): 1637-44, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15953713

RESUMEN

Kahalalide F (KF) is a small natural peptide that showed activity in vitro and in vivo. The dose-limiting toxicity in clinical trials was transaminitis. We investigated the cytotoxicity of KF in cell lines from breast, ovary, prostate and colon cancers, but focused on hepatoma cell lines, performing mechanistic studies in HepG2 (IC50 = 0.3 microM) and PLC/PRF/5C (IC50 = 5 microM). Following KF exposure, HepG2 cells demonstrated profound ATP depletion, associated with cell swelling and cell blebbing, and increased permeability to propidium iodide (PI), annexin V (AV) and release of lactate dehydrogenase (LDH). PLC/PRF/5C cells retained their cell structure, but were permeable to PI and, following exposure to high concentrations of KF, to AV. The pattern of cell permeability is similar to maitotoxin, another small cytotoxic peptide, but the differential effects on the cell membrane induced by KF in HepG2 and PLC/PRF/5C suggest specific interactions with membranes or proteins. This could lead to better drug design aimed at exploiting the potential for cell selectivity.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Depsipéptidos/farmacología , Neoplasias Hepáticas/metabolismo , Adenosina Trifosfato/deficiencia , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Depsipéptidos/farmacocinética , Humanos , Permeabilidad/efectos de los fármacos
13.
Cancer Chemother Pharmacol ; 55(6): 577-83, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15726367

RESUMEN

Ruthenium(II) organometallic complexes form monofunctional adducts with guanine in DNA in vitro and have a cytotoxic anticancer activity spectrum in preclinical models suggesting lack of cross-resistance with cisplatin. The primary cytotoxic lesion remains to be identified but the downstream mechanism of action is nevertheless of interest. Using isogenic derivatives of the HCT116 colorectal cancer cell line, we investigated the role of p53, p21/WAF1 and Bax in the cellular response to the novel ruthenium(II) organometallic complex RM175, [(eta(6)-C(6)H(5)C(6)H(5))RuCl (H(2)NCH(2)CH(2)NH(2)-N,N')](+) PF(6)(-). Western blotting demonstrated dose-dependent accumulation of p53, Bax and p21/WAF1 within 48 h of the start of RM175 treatment in wild-type HCT116 cells. HCT116 wild-type and Bax-null cells arrested in the G(1) and G(2) phases of the cell cycle. This pattern of cell cycle arrest was not observed in p53-null or in p21/WAF1-null cells. Following RM175 treatment, HCT116 wild-type and p21/WAF1 null cells underwent a dose-dependent induction of apoptosis (Annexin-V and sub-G(1) apoptosis assays). This apoptotic response was not observed in p53-null or Bax-null cells. In short-term sulphorhodamine B assays, the IC(50) for RM175 was 16 microM for p53-null HCT116, and 8 microM for wild-type cells (P<0.05). However, the sensitivity to RM175 in clonogenic assays at 16 days was independent of p53 status. These results identify determinants of the short-term in vitro response to RM175 demonstrating a role for p53 and p21/WAF1 in the growth arrest and for p53 and Bax in the apoptotic response. The mechanism of p53-independent suppression of long-term clonogenicity remains to be determined.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Ciclo Celular/metabolismo , Compuestos Organometálicos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Rutenio/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Supervivencia Celular/efectos de los fármacos , Clonación Molecular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HCT116 , Humanos , Immunoblotting , Concentración 50 Inhibidora , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteína p53 Supresora de Tumor/genética , Proteína X Asociada a bcl-2
14.
Eur J Cancer ; 40(12): 1872-7, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15288289

RESUMEN

BBR3464, a novel tri-nuclear platinum complex, forms long-range DNA adducts and is highly potent when compared with cisplatin in vitro. Preclinical studies demonstrated activity in cisplatin-resistant tumours and tumours with mutated p53 status. Phase I & II clinical studies gave preliminary indications of activity in melanoma, pancreatic, lung and ovarian cancers. The aim of this study was to determine the efficacy and confirm the toxicity of BBR3464 when given either as first- or second-line treatment for advanced disease in patients with gastric and gastro-oesphageal adenocarcinoma. Two multicentre, open label, Gehan design studies were conducted; one study used BBR3464 as first-line and the other as second-line treatment for metastatic or locally advanced disease. Nineteen first-line and 26 second-line patients were enrolled receiving a total of 74 and 53 infusions, respectively. Initially, seven patients in the second-line study received BBR3464 using the planned schedule of 1.1 mg/m2 every 4 weeks; however, 5 of these patients experienced dose-limiting grade 3 or 4 febrile neutropenia; subsequent patients in both studies were treated using the modified schedule of 0.9 mg/m2, every 21 days. In 1 of 17 evaluable, previously untreated patients, regression of multiple skin lesions was noted with stabilisation of lung metastases and maxillary sinus mass, lasting 155 days. In the first-line study, the median time to progression was 85 days [95% Confidence Interval (CI): 42, 127] (2.8 months) and in the second-line study, the median time to progression was 71 days [95% CI: 42, 109] and 38 days [95% CI: 32, 73] in the 1.1 and 0.9 mg/m2 dose level groups, respectively. Toxicity data were available for 45 patients. Neutropenia was the main toxicity seen (G3: 40%, G4: 40%). Febrile neutropenia was observed in six patients (15%) treated with 0.9 mg/m2 compared with five patients (71%) treated with 1.1 mg/m2 BBR3464. Other drug-related toxicities (G3/4) included: anaemia, thrombocytopenia, nausea, vomiting, diarrhoea, mucositis and fatigue. Diarrhoea and nausea/ vomiting were adequately controlled by the use of loperamide and antiemetics, respectively. Recruitment to the second-line study was closed early due to the poor response rate (1/17 evaluable, 6%; 95% CI: 1%, 27%) and short time to progression noted in the first-line study. Further studies with BBR3464 in this tumour type are not recommended.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Neoplasias Esofágicas/tratamiento farmacológico , Compuestos Organoplatinos/uso terapéutico , Neoplasias Gástricas/tratamiento farmacológico , Adulto , Anciano , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Persona de Mediana Edad
15.
Br J Cancer ; 90(4): 800-4, 2004 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-14970856

RESUMEN

BB-3644 is an oral, broad-spectrum matrix metalloproteinase inhibitor (MMPI) structurally related to marimastat and BB-94. It is also >10-fold more active than marimastat in inhibiting the processing of cell-bound TNF-alpha. Preclinical studies suggested a favourable toxicity profile when compared to marimastat, and therefore it was selected for clinical evaluation. Patients with advanced solid tumours against which established treatments had failed, or for which no satisfactory treatment exists and of good performance status, were eligible. Treatment consisted of twice daily (bd) oral BB-3644 for 84 days. The initial dose was 5 mg bd, and subsequent cohorts were treated with 10, 20 and 30 mg bd. In all, 22 patients were enrolled. The dose-limiting toxicity (DLT) was musculoskeletal pain. For 28 days of treatment with BB-3644, 20 mg bd was the maximum tolerated dose (MTD), as at 30 mg bd, six of nine patients developed significant musculoskeletal toxicity by day 28. Following chronic oral dosing (>28 days) with BB-3644, three of five patients treated at 10 mg bd developed musculoskeletal DLT by day 84, defining the MTD as 5 mg bd. As dose-limiting musculoskeletal toxicity was encountered at doses of BB-3644 unlikely to provide an advantage over currently available MMPIs, further evaluation is not recommended.


Asunto(s)
Aminopiridinas/efectos adversos , Aminopiridinas/farmacología , Ácidos Hidroxámicos/efectos adversos , Ácidos Hidroxámicos/farmacología , Neoplasias/tratamiento farmacológico , Administración Oral , Adulto , Anciano , Aminopiridinas/administración & dosificación , Aminopiridinas/farmacocinética , Esquema de Medicación , Femenino , Humanos , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/farmacocinética , Masculino , Dosis Máxima Tolerada , Metaloendopeptidasas , Persona de Mediana Edad , Enfermedades Musculoesqueléticas/inducido químicamente , Resultado del Tratamiento
16.
Br J Cancer ; 86(10): 1652-7, 2002 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-12085218

RESUMEN

Ruthenium complexes offer the potential of reduced toxicity, a novel mechanism of action, non-cross resistance and a different spectrum of activity compared to platinum containing compounds. Thirteen novel ruthenium(II) organometallic arene complexes have been evaluated for activity (in vitro and in vivo) in models of human ovarian cancer, and cross-resistance profiles established in cisplatin and multi-drug-resistant variants. A broad range of IC50 values was obtained (0.5 to >100 microM) in A2780 parental cells with two compounds (RM175 and HC29) equipotent to carboplatin (6 microM), and the most active compound (HC11) equipotent to cisplatin (0.6 microM). Stable bi-dentate chelating ligands (ethylenediamine), a more hydrophobic arene ligand (tetrahydroanthracene) and a single ligand exchange centre (chloride) were associated with increased activity. None of the six active ruthenium(II) compounds were cross-resistant in the A2780cis cell line, demonstrated to be 10-fold resistant to cisplatin/carboplatin by a mechanism involving, at least in part, silencing of MLH1 protein expression via methylation. Varying degrees of cross-resistance were observed in the P-170 glycoprotein overexpressing multi-drug-resistant cell line 2780AD that could be reversed by co-treatment with verapamil. In vivo activity was established with RM175 in the A2780 xenograft together with non-cross-resistance in the A2780cis xenograft and a lack of activity in the 2780AD xenograft. High activity coupled to non cross-resistance in cisplatin resistant models merit further development of this novel group of anticancer compounds.


Asunto(s)
Antineoplásicos/uso terapéutico , Azacitidina/análogos & derivados , Compuestos Organometálicos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Compuestos de Rutenio/uso terapéutico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Antimetabolitos Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Azacitidina/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Carboplatino/farmacología , Cisplatino/farmacología , Decitabina , Diseño de Fármacos , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Ácido Edético/farmacología , Femenino , Humanos , Ligandos , Ratones , Ratones Desnudos , Proteínas de Neoplasias/metabolismo , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacología , Neoplasias Ováricas/patología , Compuestos de Rutenio/química , Compuestos de Rutenio/farmacología , Relación Estructura-Actividad , Verapamilo/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Pathol ; 197(1): 128-35, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12081197

RESUMEN

Cyclin D1 protein overexpression is commonly found in colorectal carcinomas (CRCs) and is associated with a poorer prognosis, but the mechanism underlying overexpression remains uncertain. Both dysregulation of beta-catenin protein expression and k-ras mutation have recently been shown to promote cyclin D1 expression in human in vitro and rodent in vivo studies. In this study, 53 sporadic CRCs were examined by immunohistochemistry for cyclin D1 and beta-catenin protein expression, and with PCR and direct DNA sequencing for k-ras gene status. The study also addressed whether cyclin Dl overexpression might associate with poorer prognosis because of a relationship with poorer response to 5-fluorouracil (5FU) chemotherapy. Cyclin D1 overexpression was demonstrated in 34/53 (64%) CRCs, was significantly associated with higher Dukes' stage, and was particularly prominent at the invasive edges of carcinomas. Furthermore, cyclin D1 overexpression was always and only seen in association with nuclear expression of beta-catenin. There were no significant associations between cyclin D1 overexpression and k-ras mutation or response to 5FU. Amongst 17 microsatellite unstable CRCs, a smaller proportion of tumours showed cyclin D1 overexpression (18%), but again cyclin D1 overexpression was only seen in cases showing nuclear beta-catenin expression. In conclusion, beta-catenin protein dysregulation, but not k-ras mutation, appears to be required for cyclin D1 overexpression in colorectal carcinoma in vivo.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/metabolismo , Ciclina D1/metabolismo , Proteínas del Citoesqueleto/metabolismo , Genes ras , Mutación , Transactivadores , Adulto , Anciano , Antimetabolitos Antineoplásicos/uso terapéutico , División Celular , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Femenino , Fluorouracilo/uso terapéutico , Humanos , Masculino , Repeticiones de Microsatélite , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Pronóstico , Resultado del Tratamiento , beta Catenina
18.
Br J Cancer ; 85(12): 1937-43, 2001 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-11747337

RESUMEN

Thymidylate synthase (TS) is a key enzyme in DNA synthesis and is inhibited by metabolites of the chemotherapeutic agent 5-fluorouracil (5FU). Nuclear expression of TS in human tissue in vivo has not been characterised and its clinicopathological correlates in malignancy are unknown. 52 cases of primary colorectal carcinoma (CRC) and 24 cases of matched metastatic carcinoma were studied immunohistochemically using the monoclonal antibody TS106. The degree of nuclear TS immunostaining correlated closely with levels of TS mRNA expression amongst 10 CRCs studied. Strong nuclear immunostaining was seen in normal basal crypt colonocytes and germinal centre cells, and in a varying proportion of adenocarcinoma cells. Amongst the primary carcinomas, higher TS nuclear expression was associated with prominent extracellular mucin production and right-sided location. Higher TS nuclear expression also showed a significant association with poorer response to protracted venous infusional 5FU therapy. There was no clear association between TS nuclear expression and Ki67 or p53 expression assessed immunohistochemically. There was a strong positive correlation between TS nuclear expression in primary and metastatic CRC but the latter generally showed higher expression than matched primary tumour tissue. These findings confirm the nuclear expression of TS protein in human cells in vivo and provide new insight into how such expression may relate to the behaviour of CRCs.


Asunto(s)
Adenocarcinoma/genética , Antimetabolitos Antineoplásicos/uso terapéutico , Núcleo Celular/enzimología , Neoplasias Colorrectales/genética , Inhibidores Enzimáticos/uso terapéutico , Fluorouracilo/uso terapéutico , Proteínas de Neoplasias/biosíntesis , Timidilato Sintasa/biosíntesis , Proteína p53 Supresora de Tumor/biosíntesis , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/enzimología , Adenocarcinoma/mortalidad , Adulto , Anciano , Antimetabolitos Antineoplásicos/farmacocinética , Diferenciación Celular , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/mortalidad , Resistencia a Antineoplásicos/genética , Inducción Enzimática , Inhibidores Enzimáticos/farmacocinética , Femenino , Fluorouracilo/farmacocinética , Perfilación de la Expresión Génica , Genes p53 , Humanos , Antígeno Ki-67/biosíntesis , Antígeno Ki-67/genética , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , Reproducibilidad de los Resultados , Análisis de Supervivencia , Timidilato Sintasa/genética , Resultado del Tratamiento
19.
J Med Chem ; 44(22): 3616-21, 2001 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-11606126

RESUMEN

Inhibition of the growth of the human ovarian cancer cell line A2780 by organometallic ruthenium(II) complexes of the type [(eta(6)-arene)Ru(X)(Y)(Z)], where arene is benzene or substituted benzene, X, Y, and Z are halide, acetonitrile, or isonicotinamide, or X,Y is ethylenediamine (en) or N-ethylethylenediamine, has been investigated. The X-ray crystal structures of the complexes [(eta(6)-p-cymene)Ru(en)Cl]PF(6) (5), [(eta(6)-p-cymene)RuCl(2)(isonicotinamide)] (7), and [(eta(6)-biphenyl)Ru(en)Cl]PF(6) (9) are reported. They have "piano stool" geometries with eta(6) coordination of the arene ligand. Complexes with X,Y as a chelated en ligand and Z as a monofunctional leaving group had the highest activity. Complexes 5, 6 (the iodo analogue of 5), 9, and 10 (ethylethylenediamine analogue of 9) were as active as carboplatin. Hydrolysis of the reactive Ru-Cl bond in complex 5 was detected by HPLC but was suppressed by the addition of chloride ions. Complex 5 binds strongly and selectively to G bases on DNA oligonucleotides to form monofunctional adducts. No inhibition of topoisomerase I or II by complexes 5, 6, or 9 was detected. These chelated Ru(II) arene complexes have potential as novel metal-based anticancer agents with a mechanism of action different from that of the Ru(III) complex currently on clinical trial.


Asunto(s)
Antineoplásicos/síntesis química , Compuestos Organometálicos/síntesis química , Rutenio , Antineoplásicos/química , Antineoplásicos/farmacología , Cristalografía por Rayos X , Aductos de ADN/química , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Estructura Molecular , Oligonucleótidos/química , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacología , Relación Estructura-Actividad , Inhibidores de Topoisomerasa I , Inhibidores de Topoisomerasa II , Células Tumorales Cultivadas
20.
Clin Cancer Res ; 7(10): 3071-8, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11595697

RESUMEN

PURPOSE: Arg-D-Trp-NmePhe-D-Trp-Leu-Met-NH(2) (Antagonist G), a substance P (SP 6-11) analogue, inhibits mitogenesis stimulated by a broad spectrum of neuropeptides and has demonstrated antitumor activity in vitro and in vivo with IC(50) concentrations of 10-20 microM in small cell lung cancer and other cell lines. Because neuropeptides are part of complex neurohumoral pathways, we have sought to develop novel pharmacodynamic approaches as part of the early clinical development of this potential anticancer drug. EXPERIMENTAL DESIGN: A Phase I trial was performed in two stages. In stage 1, Antagonist G was administered at 3- week intervals using an accelerated dose-escalation strategy until the target maximum plasma concentration (C(max)) of 10 microM was achieved. In stage 2, dose intensity was increased to weekly, and the inhibitory effect of i.v. Antagonist G was assessed by forearm blood flow (FBF) using SP as a vasodilator, as measured by venous plethysmography. RESULTS: In stage 1, dose was escalated from 2 to 300 mg/m(2) in 12 dose levels using only 15 patients. In stage 2, nine patients were entered at three dose levels (300, 350, and 400 mg/m(2)) and a C(max) of 45 microM was achieved. Facial flushing was the only consistent toxicity but was not dose limiting. FBF studies demonstrated that Antagonist G consistently inhibited the vasodilatory effects of SP (mean, 62 +/- 2% inhibition). CONCLUSIONS: Antagonist G can be safely administered up to 400 mg/m(2), achieving C(max)s >20 microM by weekly 6-h i.v. infusion. FBF studies in patients demonstrated that Antagonist G inhibits SP vasodilatory effects in vivo at these doses in the absence of dose-limiting toxicity.


Asunto(s)
Antineoplásicos/uso terapéutico , Antebrazo/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Oligopéptidos/uso terapéutico , Vasodilatadores/farmacología , Adulto , Anciano , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Arteria Braquial/efectos de los fármacos , Bradiquinina/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Rubor/inducido químicamente , Humanos , Bombas de Infusión , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Neoplasias/fisiopatología , Neuropéptidos/antagonistas & inhibidores , Oligopéptidos/sangre , Oligopéptidos/farmacocinética , Flujo Sanguíneo Regional/efectos de los fármacos , Sustancia P/farmacología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA