Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Bioorg Med Chem ; 19(22): 6949-65, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21982796

RESUMO

The cyclin-dependent kinase (CDK) inhibitor seliciclib (1, CYC202) is in phase II clinical development for the treatment of cancer. Here we describe the synthesis of novel purines with greater solubility, lower metabolic clearance, and enhanced potency versus CDKs. These compounds exhibit novel selectivity profiles versus CDK isoforms. Compound αSßR-21 inhibits CDK2/cyclin E with IC(50)=30 nM, CDK7-cyclin H with IC(50)=1.3 µM, and CDK9-cyclinT with IC(50)=0.11 µM; it (CCT68127) inhibits growth of HCT116 colon cancer cells in vitro with GI(50)=0.7 µM; and shows antitumour activity when dosed p.o. at 50mg/kg to mice bearing HCT116 solid human tumour xenografts.


Assuntos
Adenosina/análogos & derivados , Quinases Ciclina-Dependentes/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Purinas/química , Adenosina/síntese química , Adenosina/química , Adenosina/farmacocinética , Adenosina/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/farmacologia , Purinas/síntese química , Purinas/farmacocinética , Purinas/farmacologia , Roscovitina , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Biochem J ; 429(3): 565-72, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20497126

RESUMO

VEGF (vascular endothelial growth factor) plays an essential role in angiogenesis during development and in disease largely mediated by signalling events initiated by binding of VEGF to its receptor, VEGFR2 (VEGF receptor 2)/KDR (kinase insert domain receptor). Recent studies indicate that VEGF activates PKD (protein kinase D) in endothelial cells to regulate a variety of cellular functions, including signalling events, proliferation, migration and angiogenesis. To better understand the role of PKD in VEGF-mediated endothelial function, we characterized the effects of a novel pyrazine benzamide PKD inhibitor CRT5 in HUVECs (human umbilical vein endothelial cells). The activity of the isoforms PKD1 and PKD2 were blocked by this inhibitor as indicated by reduced phosphorylation, at Ser916 and Ser876 respectively, after VEGF stimulation. The VEGF-induced phosphorylation of three PKD substrates, histone deacetylase 5, CREB (cAMP-response-element-binding protein) and HSP27 (heat-shock protein 27) at Ser82, was also inhibited by CRT5. In contrast, CRT6, an inactive analogue of CRT5, had no effect on PKD or HSP27 Ser82 phosphorylation. Furthermore, phosphorylation of HSP27 at Ser78, which occurs solely via the p38 MAPK (mitogen-activated protein kinase) pathway, was also unaffected by CRT5. In vitro kinase assays show that CRT5 did not significantly inhibit several PKC isoforms expressed in endothelial cells. CRT5 also decreased VEGF-induced endothelial migration, proliferation and tubulogenesis, similar to effects seen when the cells were transfected with PKD siRNA (small interfering RNA). CRT5, a novel specific PKD inhibitor, will greatly facilitate the study of the role of PKD signalling mechanisms in angiogenesis.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/enzimologia , Humanos , Neovascularização Fisiológica/efeitos dos fármacos , Fosforilação , Especificidade por Substrato , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
3.
Mol Cancer Ther ; 9(5): 1136-46, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20442301

RESUMO

Protein kinase D (PKD) family members are increasingly implicated in multiple normal and abnormal biological functions, including signaling pathways that promote mitogenesis in pancreatic cancer. However, nothing is known about the effects of targeting PKD in pancreatic cancer. Our PKD inhibitor discovery program identified CRT0066101 as a specific inhibitor of all PKD isoforms. The aim of our study was to determine the effects of CRT0066101 in pancreatic cancer. Initially, we showed that autophosphorylated PKD1 and PKD2 (activated PKD1/2) are significantly upregulated in pancreatic cancer and that PKD1/2 are expressed in multiple pancreatic cancer cell lines. Using Panc-1 as a model system, we showed that CRT0066101 reduced bromodeoxyuridine incorporation; increased apoptosis; blocked neurotensin-induced PKD1/2 activation; reduced neurotensin-induced, PKD-mediated Hsp27 phosphorylation; attenuated PKD1-mediated NF-kappaB activation; and abrogated the expression of NF-kappaB-dependent proliferative and prosurvival proteins. We showed that CRT0066101 given orally (80 mg/kg/d) for 24 days significantly abrogated pancreatic cancer growth in Panc-1 subcutaneous xenograft model. Activated PKD1/2 expression in the treated tumor explants was significantly inhibited with peak tumor concentration (12 micromol/L) of CRT0066101 achieved within 2 hours after oral administration. Further, we showed that CRT0066101 given orally (80 mg/kg/d) for 21 days in Panc-1 orthotopic model potently blocked tumor growth in vivo. CRT0066101 significantly reduced Ki-67-positive proliferation index (P < 0.01), increased terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling-positive apoptotic cells (P < 0.05), and abrogated the expression of NF-kappaB-dependent proteins including cyclin D1, survivin, and cIAP-1. Our results show for the first time that a PKD-specific small-molecule inhibitor CRT0066101 blocks pancreatic cancer growth in vivo and show that PKD is a novel therapeutic target in pancreatic cancer.


Assuntos
Carcinoma Ductal Pancreático/patologia , Proliferação de Células/efeitos dos fármacos , Neoplasias Pancreáticas/patologia , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma Ductal Pancreático/tratamento farmacológico , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos Nus , Peso Molecular , Neoplasias Pancreáticas/tratamento farmacológico , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Biochem Pharmacol ; 78(2): 115-22, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19475728

RESUMO

The ability of two structurally diverse telomeric G-quadruplex-binding compounds to synergise the action of cis-platin has been investigated in two cancer cell lines. One compound is a trisubstituted acridine compound AS1410, a close analogue of BRACO-19, and the other is a non-polycyclic compound synthesised using click chemistry and containing two triazole rings. Both compounds produce growth arrest at sub-cytotoxic concentrations in the two cell lines (MCF7 and A549), with behaviour consistent with telomere targeting mechanisms. Synergistic behaviour was observed in both cell lines with both compounds in combination with cis-platin, but only when the ratio of AS1410:cis-platin is >1. In vivo tumour xenograft studies with the A549 lung cancer model and the trisubstituted acridine compound AS1410 showed only a modest anti-tumour effect when administered alone, but produced rapid and highly significant decreases in tumour volume when administered in combination with cis-platin.


Assuntos
Antineoplásicos/administração & dosagem , Cisplatino/administração & dosagem , Quadruplex G , Inibidores do Crescimento/administração & dosagem , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Animais , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Cisplatino/farmacologia , Sinergismo Farmacológico , Feminino , Inibidores do Crescimento/farmacologia , Humanos , Camundongos , Camundongos Nus , Fatores de Tempo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
5.
J Med Chem ; 51(7): 2254-60, 2008 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-18338842

RESUMO

The chemistry and biology of acetyl-protected spermidine-bridged dinuclear platinum complexes [{ trans-PtCl(NH 3) 2] 2-mu-NH 2(CH 2) 3N(COR)(CH 2) 4NH 2]X 2 (R = H, X = Cl (1,1/t,t-spermidine, BBR3571); R = CH 3 , X = Cl ( 2); R = CH 2 Cl, X = ClO 4 ( 3); R = CF 3 , X = Cl ( 4)) are compared with their carbamate analogues. The compounds are potential prodrugs for the parent compound 1, a highly potent antitumor agent. At pH 6-8 hydrolysis of the blocking group with the release of the "parent" protonated species follows the order 4 > 3 >> 2. For 4, rate constants for the deprotection increase in this pH range. The DNA binding profile of 4 is similar to the Boc derivative, confirming the central influence of charge on DNA binding properties. The differences in cytotoxicity for the protected compounds in ovarian carcinoma cell lines sensitive and resistant to cisplatin cannot completely be explained by spontaneous release of 1,1/t,t-spermidine at physiological pH. Inherent cytotoxicity and cell line specificity may contribute to the observed behavior. The properties of the compounds present them also as possible "second-generation" analogues of the clinically relevant trinuclear complex [{ trans-PtCl(NH 3) 2} 2-mu- trans-Pt(NH 3) 2(NH 2(CH 2) 6NH 2) 2](NO 3) 4, ( 8, BBR3464).


Assuntos
Amidas/química , DNA/efeitos dos fármacos , Compostos Organoplatínicos/síntese química , Compostos Organoplatínicos/farmacologia , Pró-Fármacos/síntese química , Pró-Fármacos/farmacologia , Espermidina/química , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , DNA/química , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Concentração de Íons de Hidrogênio , Hidrólise , Dados de Sequência Molecular , Estrutura Molecular , Compostos Organoplatínicos/química , Pró-Fármacos/química , Sensibilidade e Especificidade , Estereoisomerismo , Fatores de Tempo
6.
Clin Cancer Res ; 13(17): 4960-3, 2007 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-17785545

RESUMO

The maintenance of telomeric DNA underlies the ability of tumors to possess unlimited replicative potential, one of the hallmarks of cancer. Telomere length and structure are maintained by the reverse transcriptase telomerase and a multiprotein telomere complex termed shelterin. Telomerase activity is elevated in the vast majority of tumors, and telomeres are critically shortened in tumors versus normal tissues, thus providing a compelling rationale to target the telomerase/telomere pathway for broad-spectrum cancer therapy. This strategy is supported by a variety of genetic-based target validation studies. Both telomerase inhibitors and telomere interactive molecules have shown stand-alone antitumor activity at nontoxic doses against a variety of human tumor xenografts in mice. These translational advances have resulted in the first antitelomerase agent, the oligonucleotide-based GRN163L targeting the telomerase RNA template, entering clinical evaluation. Additional translational approaches, such as targeting telomeres using G-quadruplex ligands, should result in antitelomere agents, such as RHPS4, entering the clinic in the near future. These prototype trials will be extremely informative in determining the role of the telomerase/telomere pathway in clinical oncology and, moreover, whether drugs targeting the unlimited replicative potential of cancer will find a place in cancer chemotherapy.


Assuntos
Neoplasias/tratamento farmacológico , Telomerase/antagonistas & inibidores , Telômero , Animais , Humanos , Neoplasias/enzimologia , Neoplasias/etiologia , Neoplasias/genética , Telomerase/fisiologia
7.
Nat Rev Cancer ; 7(8): 573-84, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17625587

RESUMO

The accidental discovery of the anticancer properties of cisplatin and its clinical introduction in the 1970s represent a major landmark in the history of successful anticancer drugs. Although carboplatin--a second-generation analogue that is safer but shows a similar spectrum of activity to cisplatin--was introduced in the 1980s, the pace of further improvements slowed for many years. However, in the past several years interest in platinum drugs has increased. Key developments include the elucidation of mechanisms of tumour resistance to these drugs, the introduction of new platinum-based agents (oxaliplatin, satraplatin and picoplatin), and clinical combination studies using platinum drugs with resistance modulators or new molecularly targeted drugs.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Compostos de Platina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Humanos
8.
Expert Opin Investig Drugs ; 16(7): 1009-21, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17594186

RESUMO

The three platinum-containing drugs that have been thus far approved by the FDA - cisplatin, carboplatin and oxaliplatin - have had a significant effect in the treatment of patients with some malignancies such as testicular, ovarian and colorectal cancer. However, much more remains to be achieved to widen the therapeutic use of this important class of drug, either via further analogue development or by judicious use of combining the existing drugs with new molecularly targeted agents. Two analogues arising from an academic (Institute of Cancer Research)/pharmaceutical (Johnson Matthey/AnorMed) collaboration - satraplatin (JM-216) and picoplatin (JM-/AMD-473) - have recently shown promising clinical activity; satraplatin (an orally available drug) in hormone-refractory prostate cancer and picoplatin in small-cell lung cancer. There have also been advances in delivery vehicles for platinum drugs (e.g., the diaminocyclohexane [DACH]-based AP-5346 and aroplatin/liposomal cis-bis-neodecanoato-trans-(R,R)-1,2-diaminocyclohexane platinum (II) [L-NDDP] are in early clinical development). Platinum-based drugs have also been successfully combined with molecularly targeted drugs (e.g., the recent approval of the vascular endothelial growth factor monoclonal antibody bevacizumab with carboplatin and paclitaxel in patients with NSCLC).


Assuntos
Drogas em Investigação , Neoplasias/tratamento farmacológico , Compostos Organoplatínicos/administração & dosagem , Compostos de Platina/administração & dosagem , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Ensaios Clínicos Fase II como Assunto , Ensaios Clínicos Fase III como Assunto , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Masculino , Dose Máxima Tolerável , Neoplasias/mortalidade , Neoplasias/patologia , Compostos Organoplatínicos/efeitos adversos , Compostos Organoplatínicos/farmacologia , Compostos de Platina/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Sensibilidade e Especificidade , Resultado do Tratamento
9.
Cancer Res ; 67(12): 5840-50, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17575152

RESUMO

Extensive evidence implicates activation of the lipid phosphatidylinositide 3-kinase (PI3K) pathway in the genesis and progression of various human cancers. PI3K inhibitors thus have considerable potential as molecular cancer therapeutics. Here, we detail the pharmacologic properties of a prototype of a new series of inhibitors of class I PI3K. PI103 is a potent inhibitor with low IC50 values against recombinant PI3K isoforms p110alpha (2 nmol/L), p110beta (3 nmol/L), p110delta (3 nmol/L), and p110gamma (15 nmol/L). PI103 also inhibited TORC1 by 83.9% at 0.5 micromol/L and exhibited an IC50 of 14 nmol/L against DNA-PK. A high degree of selectivity for the PI3K family was shown by the lack of activity of PI103 in a panel of 70 protein kinases. PI103 potently inhibited proliferation and invasion of a wide variety of human cancer cells in vitro and showed biomarker modulation consistent with inhibition of PI3K signaling. PI103 was extensively metabolized, but distributed rapidly to tissues and tumors. This resulted in tumor growth delay in eight different human cancer xenograft models with various PI3K pathway abnormalities. Decreased phosphorylation of AKT was observed in U87MG gliomas, consistent with drug levels achieved. We also showed inhibition of invasion in orthotopic breast and ovarian cancer xenograft models and obtained evidence that PI103 has antiangiogenic potential. Despite its rapid in vivo metabolism, PI103 is a valuable tool compound for exploring the biological function of class I PI3K and importantly represents a lead for further optimization of this novel class of targeted molecular cancer therapeutic.


Assuntos
Antineoplásicos/farmacologia , Furanos/farmacologia , Neoplasias/tratamento farmacológico , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Inibidores de Fosfoinositídeo-3 Quinase , Piridinas/farmacologia , Pirimidinas/farmacologia , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Citometria de Fluxo , Humanos , Immunoblotting , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Biochem Pharmacol ; 74(1): 20-7, 2007 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-17466278

RESUMO

P53 gene status is implicated in the cytotoxic drug sensitivity and published research has been mostly addressed to cisplatin (CDDP) activity. Previous study in our laboratory considered p53 mutant cell lines A431 (parental) and A431/Pt (CDDP-resistant counterpart, resistance factor R.F.=2.6). For a comparison which contributes to a deeper appreciation of the process that mediates the Pt drug cellular effects, we extended our investigation to the p53 wild-type cell lines U2-OS (human osteosarcoma) and its CDDP-resistant counterpart U2-OS/Pt (R.F.=5). We compared the activity of CDDP, oxaliplatin (L-OHP) and satraplatin (JM216) whose hydrophobicity rank is JM216>L-OHP>CDDP. In U2-OS cells the three drugs accumulated similarly, while in U2-OS/Pt the most hydrophobic drugs were privileged. No significant differences in efflux were observed between sensitive and resistant cell lines. The growing of CDDP resistance seems to be overcome by increasing the hydrophobicity of the Pt agent. An almost linear trend seems to relate R.F. and drug hydrophobicity in U2-OS/Pt and A431/Pt cells. DNA platination in U2-OS as in A431 cells is at the lowest levels for L-OHP. In U2-OS cell line the IC(50) of CDDP (17.6 microM) and JM216 (88.02 microM) do not correlate with their similar levels of Pt-DNA adducts (mean value approximately 0.14 pmol Pt/microg DNA). The presence of a wild-type p53 exalts either CDDP cytotoxicity (two-fold more active in U2-OS than in A431 cells) and CDDP resistance in comparison to a p53 mutant type. The p53 status seems to not improve JM216 or L-OHP cytotoxicity in both cell lines.


Assuntos
Antineoplásicos/toxicidade , Adutos de DNA/metabolismo , Compostos Organoplatínicos/toxicidade , Osteossarcoma/tratamento farmacológico , Proteína Supressora de Tumor p53/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/toxicidade , DNA de Neoplasias/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Concentração Inibidora 50 , Mutação , Osteossarcoma/genética , Osteossarcoma/metabolismo , Oxaliplatina , Proteína Supressora de Tumor p53/metabolismo
11.
Curr Opin Mol Ther ; 9(1): 86-91, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17330406

RESUMO

Adherex Technologies Inc, under license from McGill University, is developing ADH-1, an intravenous N-cadherin antagonist that specifically disrupts the disorganized interactions between N-cadherin molecules in tumor blood vessels, as a potential anticancer compound. By May 2005, ADH-1 was in phase Ib/II clinical trials in Europe and the US, and phase II clinical trials in Canada.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Caderinas/antagonistas & inibidores , Ensaios Clínicos como Assunto , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Oligopeptídeos/farmacologia , Oligopeptídeos/uso terapêutico , Peptídeos Cíclicos/farmacologia , Peptídeos Cíclicos/uso terapêutico , Inibidores da Angiogênese/efeitos adversos , Inibidores da Angiogênese/síntese química , Inibidores da Angiogênese/farmacocinética , Animais , Humanos , Neoplasias/irrigação sanguínea , Oligopeptídeos/síntese química , Oligopeptídeos/farmacocinética , Peptídeos Cíclicos/síntese química , Peptídeos Cíclicos/farmacocinética
12.
Bioorg Med Chem Lett ; 17(8): 2293-8, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17276687

RESUMO

The design, synthesis, biophysical and biochemical evaluation is presented of a new series of benzylamino-substituted acridines as G-quadruplex binding telomerase inhibitors. Replacement of the previously reported anilino substituents by benzylamino groups results in enhanced quadruplex interaction, and for one compound, superior telomerase inhibitory activity.


Assuntos
Acridinas/síntese química , Benzilaminas/síntese química , DNA/efeitos dos fármacos , Telomerase/antagonistas & inibidores , Telômero/efeitos dos fármacos , Acridinas/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Benzilaminas/farmacologia , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Quadruplex G , Guanosina , Humanos
13.
Invest New Drugs ; 25(3): 271-6, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17203401

RESUMO

AIMS: DMXAA (AS1404), a small-molecule vascular disrupting agent that has now completed Phase II clinical trial, induces endothelial cell apoptosis, increased vascular permeability and decreased tumour blood flow in vivo. Its action is incompletely understood and we wished to develop an in vitro system to study its effects. METHODS: Human tumour cell lines developed from aggressive tumours were grown on Matrigel to simulate a tumour microenvironment. Cells were analysed by light microscopy and by gene expression profiling. RESULTS: Several cell lines formed networks when grown on Matrigel and the NZM7 melanoma cell line was chosen for further study. Addition of DMXAA at a clinically achievable concentration (30 microg/mL) prevented network formation, but co-addition of SB203580 (10 microM), a selective inhibitor of p38 MAP kinase, reversed the effect of DMXAA and restored network formation. Analysis of expression genes for endothelial and related functions showed that cells growing on Matrigel expressed a pattern similar to that of NZM7 cells growing as xenografts in vivo but different from that of cells grown on standard tissue culture plates. Addition of DMXAA resulted in the inhibition of expression of several genes including the transcriptional activator Ets1 and matrix metalloproteinase-2 (MMP2), but co-addition of SB203580 did not reverse these effects of DMXAA on gene expression. CONCLUSION: The results suggest that p38 MAP kinase plays an important role in the action of DMXAA and that growth of tumour cells on Matrigel provides a promising model for further studies on the action of this drug.


Assuntos
Inibidores da Angiogênese/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melanoma/enzimologia , Xantonas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Colágeno/metabolismo , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imidazóis/farmacologia , Laminina/metabolismo , Melanoma/irrigação sanguínea , Melanoma/genética , Melanoma/patologia , Camundongos , Camundongos Nus , Inibidores de Proteínas Quinases/farmacologia , Proteoglicanas/metabolismo , Piridinas/farmacologia , RNA Mensageiro/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
14.
Mol Cancer Ther ; 5(12): 2957-62, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17172400

RESUMO

Aptamers, also termed as decoys or "chemical antibodies," represent an emerging class of therapeutics. They are short DNA or RNA oligonucleotides or peptides that assume a specific and stable three-dimensional shape in vivo, thereby providing specific tight binding to protein targets. In some cases and as opposed to antisense oligonucleotides, effects can be mediated against extracellular targets, thereby preventing a need for intracellular transportation. The first aptamer approved for use in man is a RNA-based molecule (Macugen, pegaptanib) that is administered locally (intravitreally) to treat age-related macular degeneration by targeting vascular endothelial growth factor. The most advanced aptamer in the cancer setting is AS1411, formerly known as AGRO100, which is being administered systemically in clinical trials. AS1411 is a 26-mer unmodified guanosine-rich oligonucleotide, which induces growth inhibition in vitro, and has shown activity against human tumor xenografts in vivo. The mechanism underlying its antiproliferative effects in cancer cells seems to involve initial binding to cell surface nucleolin and internalization, leading to an inhibition of DNA replication. In contrast to other unmodified oligonucleotides, AS1411 is relatively stable in serum-containing medium, probably as a result of the formation of dimers and a quartet structure. In a dose escalation phase I study in patients with advanced solid tumors, doses up to 10 mg/kg/d (using a four or seven continuous infusion regime) have been studied. Promising signs of activity have been reported (multiple cases of stable disease and one near complete response in a patient with renal cancer) in the absence of any significant adverse effects. Further trials are ongoing in renal and non-small cell lung cancers. In preclinical studies, additional aptamers have been described against several cancer targets, such as tenascin-C, the transcription factor signal transducer and activator of transcription 3, and antiapoptotic and Ku proteins.


Assuntos
Aptâmeros de Nucleotídeos/farmacologia , Aptâmeros de Peptídeos/farmacologia , Neoplasias/tratamento farmacológico , Animais , Aptâmeros de Nucleotídeos/uso terapêutico , Aptâmeros de Peptídeos/uso terapêutico , Humanos , Neoplasias/genética , Oligodesoxirribonucleotídeos/farmacocinética , Oligodesoxirribonucleotídeos/farmacologia , Oligodesoxirribonucleotídeos/uso terapêutico
15.
Expert Opin Investig Drugs ; 15(11): 1309-18, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17040193

RESUMO

This perspective is the second in a series of papers discussing drugs dropped from development in 2005, of which 20 were being developed for cancer. Of these 20, 60% were biological agents and included 3 cancer vaccine (e.g., Canvaxin) and 2 antisense oligonucleotide (e.g., aprinocarsen sodium). Overall, 10 cancer drugs were discontinued in Phase I, 6 in Phase II and 4 at Phase III. This paper surveys drugs discontinued in the oncology arena in 2005 by stage of development and class of compound, suggesting some reasons for their failure.


Assuntos
Antineoplásicos , Ensaios Clínicos como Assunto , Drogas em Investigação , Antineoplásicos/uso terapêutico , Drogas em Investigação/uso terapêutico , Humanos , Falha de Tratamento
16.
Oncol Res ; 16(2): 97-106, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16898270

RESUMO

The melanin biosynthetic pathway from tyrosine is a potential target for combating malignant melanoma. N-Acetyl-4-S-cysteaminylphenol 1 is a previously synthesized analogue of tyrosine that probably acts by this pathway. It interferes with cell growth and proliferation via selective oxidation in melanocytes to an oquinone that can alkylate cellular nucleophiles. We previously synthesized a range of analogues of the original lead compound 1 most of which displayed greater cytotoxicity than 1. Eighteen new analogues with the amide group reversed have now been synthesized and tested for antimelanoma activity. Most of these reverse amides showed greater cytotoxicity than N-acetyl-4-S-cysteaminylphenol towards five representative melanoma cell lines. The highest cytotoxicity was observed for the piperidine and hexamethyleneimine derivatives 7, 8, 12, 13, and 17 and the catechol 18. The most active compound, 7, had cytotoxicity comparable to cisplatin against the five melanoma cell lines. The moderate activity of 7 and 18 against SK-Mel-24 (non-tyrosinase containing) and an ovarian cell line suggests that interference with the melanin pathway may not be the only mode of action of these compounds. Assays of some of the compounds as substrates for tyrosinase showed that the catechol 18 was the best substrate and that the piperidine derivative 7 was the best substrate of the phenolic compounds synthesized.


Assuntos
Amidas/química , Antineoplásicos/síntese química , Antineoplásicos/toxicidade , Cisteamina/análogos & derivados , Melanoma/tratamento farmacológico , Melanoma/patologia , Fenóis/química , Fenóis/toxicidade , Antineoplásicos/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cisteamina/síntese química , Cisteamina/química , Cisteamina/toxicidade , Humanos , Estrutura Molecular , Fenóis/síntese química , Relação Estrutura-Atividade
17.
Neoplasia ; 8(3): 199-206, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16611413

RESUMO

The dose-dependent effects of 5,6-dimethylxanthenone-4-acetic acid (DMXAA) on rat GH3 prolactinomas were investigated in vivo. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) was used to assess tumor blood flow/permeability pretreatment and 24 hours posttreatment with 0, 100, 200, or 350 mg/kg DMXAA. DCE-MRI data were analyzed using K(trans) and the integrated area under the gadolinium time curve (IAUGC) as response biomarkers. High-performance liquid chromatography (HPLC) was used to determine the plasma concentration of the serotonin metabolite 5-hydroxyindoleacetic acid (5-HIAA) following treatment to provide an index of increased vessel permeability and vascular damage. Finally, tumor necrosis was assessed by grading hematoxylin and eosin-stained sections cut from the same tumors investigated by MRI. Both tumor K(trans) and IAUGC were significantly reduced 24 hours posttreatment with 350 mg/kg DMXAA only, with no evidence of dose response. HPLC demonstrated a significant increase in plasma 5-HIAA 24 hours posttreatment with 200 and 350 mg/kg DMXAA. Histologic analysis revealed some evidence of tumor necrosis following treatment with 100 or 200 mg/kg DMXAA, reaching significance with 350 mg/kg DMXAA. The absence of any reduction in K(trans) or IAUGC following treatment with 200 mg/kg, despite a significant increase in 5-HIAA, raises concerns about the utility of established DCE-MRI biomarkers to assess tumor response to DMXAA.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Indóis/sangue , Imageamento por Ressonância Magnética , Neovascularização Patológica/tratamento farmacológico , Neoplasias Hipofisárias/patologia , Prolactinoma/irrigação sanguínea , Xantonas/uso terapêutico , Inibidores da Angiogênese/farmacocinética , Inibidores da Angiogênese/farmacologia , Animais , Área Sob a Curva , Biomarcadores , Permeabilidade Capilar , Linhagem Celular Tumoral/transplante , Meios de Contraste , Relação Dose-Resposta a Droga , Feminino , Gadolínio DTPA , Necrose , Transplante de Neoplasias , Prolactinoma/tratamento farmacológico , Prolactinoma/patologia , Ratos , Ratos Endogâmicos WF , Tela Subcutânea , Xantonas/farmacocinética , Xantonas/farmacologia
19.
Inorg Chem ; 44(26): 9598-600, 2005 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-16363817

RESUMO

Charge delocalization (6+ to 8+) in "noncovalent" linear trinuclear platinum complexes produces compounds with cytotoxicity in some cases equivalent to cisplatin. The cellular uptake of a novel 8+ compound is greater than that of neutral cisplatin as well as other multinuclear Pt compounds.


Assuntos
Compostos de Platina/síntese química , Compostos de Platina/toxicidade , Linhagem Celular Tumoral , Humanos , Estrutura Molecular , Compostos de Platina/química
20.
Clin Cancer Res ; 11(19 Pt 1): 7023-32, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16203796

RESUMO

PURPOSE: To establish the pharmacokinetic and pharmacodynamic profile of the heat shock protein 90 (HSP90) inhibitor 17-allylamino, 17-demethoxygeldanamycin (17-AAG) in ovarian cancer xenograft models. EXPERIMENTAL DESIGN: The effects of 17-AAG on growth inhibition and the expression of pharmacodynamic biomarkers c-RAF-1, CDK4, and HSP70 were studied in human ovarian cancer cell lines A2780 and CH1. Corresponding experiments were conducted with established tumor xenografts. The variability and specificity of pharmacodynamic markers in human peripheral blood lymphocytes (PBL) were studied. RESULTS: The IC50 values of 17-AAG in A2780 and CH1 cells were 18.3 nmol/L (SD, 2.3) and 410.1 nmol/L (SD, 9.4), respectively. Pharmacodynamic changes indicative of HSP90 inhibition were demonstrable at greater than or equal the IC50 concentration in both cell lines. Xenograft experiments confirmed tumor growth inhibition in vivo. Peak concentrations of 17-AAG achieved in A2780 and CH1 tumors were 15.6 and 16.5 micromol/L, respectively, and there was no significant difference between day 1 and 11 pharmacokinetic profiles. Reversible changes in pharmacodynamic biomarkers were shown in tumor and murine PBLs in both xenograft models. Expression of pharmacodynamic markers varied between human PBLs from different human volunteers but not within the same individual. Pharmacodynamic biomarker changes consistent with HSP90 inhibition were shown in human PBLs exposed ex vivo to 17-AAG but not to selected cytotoxic drugs. CONCLUSION: Pharmacokinetic-pharmacodynamic relationships were established for 17-AAG. This information formed the basis of a pharmacokinetic-pharmacodynamic-driven phase I trial.


Assuntos
Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Rifabutina/análogos & derivados , Animais , Antineoplásicos/farmacologia , Benzoquinonas , Biomarcadores/metabolismo , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , Quinase 4 Dependente de Ciclina/metabolismo , Relação Dose-Resposta a Droga , Feminino , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Concentração Inibidora 50 , Lactamas Macrocíclicas , Linfócitos/citologia , Camundongos , Transplante de Neoplasias , Proteínas Proto-Oncogênicas c-raf/metabolismo , Rifabutina/farmacocinética , Rifabutina/farmacologia , Fatores de Tempo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...