Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Biol Ther ; 23(1): 117-126, 2022 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-35129069

RESUMO

Mitochondria are key tumor drivers, but their suitability as a therapeutic target is unknown. Here, we report on the preclinical characterization of Gamitrinib (GA mitochondrial matrix inhibitor), a first-in-class anticancer agent that couples the Heat Shock Protein-90 (Hsp90) inhibitor 17-allylamino-geldanamycin (17-AAG) to the mitochondrial-targeting moiety, triphenylphosphonium. Formulated as a stable (≥24 weeks at -20°C) injectable suspension produced by microfluidization (<200 nm particle size), Gamitrinib (>99.5% purity) is heavily bound to plasma proteins (>99%), has intrinsic clearance from liver microsomes of 3.30 mL/min/g and minimally penetrates a Caco-2 intestinal monolayer. Compared to 17-AAG, Gamitrinib has slower clearance (85.6 ± 5.8 mL/min/kg), longer t1/2 (12.2 ± 1.55 h), mean AUC0-t of 783.1 ± 71.3 h∙ng/mL, and unique metabolism without generation of 17-AG. Concentrations of Gamitrinib that trigger tumor cell killing (IC50 ~1-4 µM) do not affect cytochrome P450 isoforms CYP1A2, CYP2A6, CYP2B6, CYP2C8 or ion channel conductance (Nav1.5, Kv4.3/KChIP2, Cav1.2, Kv1.5, KCNQ1/mink, HCN4, Kir2). Twice weekly IV administration of Gamitrinib to Sprague-Dawley rats or beagle dogs for up to 36 d is feasible. At dose levels of up to 5 (rats)- and 12 (dogs)-fold higher than therapeutically effective doses in mice (10 mg/kg), Gamitrinib treatment is unremarkable in dogs with no alterations in clinical-chemistry parameters, heart function, or tissue histology, and causes occasional inflammation at the infusion site and mild elevation of serum urea nitrogen in rats (≥10 mg/kg/dose). Therefore, targeting mitochondria for cancer therapy is feasible and well tolerated. A publicly funded, first-in-human phase I clinical trial of Gamitrinib in patients with advanced cancer is ongoing (ClinicalTrials.gov NCT04827810).


Assuntos
Antineoplásicos , Mitocôndrias , Neoplasias , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Benzoquinonas/metabolismo , Benzoquinonas/farmacologia , Células CACO-2 , Ensaios Clínicos Fase I como Assunto , Cães , Estudos de Viabilidade , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Lactamas Macrocíclicas/metabolismo , Lactamas Macrocíclicas/farmacologia , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neoplasias/tratamento farmacológico , Ratos , Ratos Sprague-Dawley
2.
Int J Toxicol ; 33(3): 162-174, 2014 05.
Artigo em Inglês | MEDLINE | ID: mdl-24801488

RESUMO

Historically, nitrogen oxides (NOx) in food, drinking water, as well as in the atmosphere have been believed to be associated with adverse health consequences. More recently, NOx have been implicated in normal homeostatic regulation, and exogenous administration has been associated with health benefits. One such potential health benefit is the prospect that inhaled nitrite will lower pulmonary blood pressure (BP) in patients with pulmonary arterial hypertension (PAH), a disease with poor prognosis due to the lack of effective treatment. To characterize potential chronic toxicity associated with inhaled AIR001 (sodium nitrite) for use in the treatment of PAH, 26-week exposures to AIR001 were carried out by inhalation administration in rats and by intravenous infusion in dogs. The studies revealed that methemoglobinemia was the primary adverse effect in both species. Methemoglobin levels less than 40% were well tolerated in both species, while levels greater than 50% methemoglobin caused death in some rats. Additionally, a decrease in systemic BP was also observed with inhaled AIR001 exposure in dogs. These acute secondary and exaggerated pharmacological effects occurred daily throughout the 26-week treatment period. Chronic exposure did not alter the magnitude of either methemoglobinemia or hypotension or result in additional toxicity or compensatory responses. Based on the exposure levels that produced these pharmacodynamic responses in animals, relative to those measured in early clinical studies, it appears that an adequate margin of safety exists to support the continued clinical development of inhaled AIR001.


Assuntos
Anti-Hipertensivos/efeitos adversos , Drogas em Investigação/efeitos adversos , Cavidade Nasal/efeitos dos fármacos , Mucosa Nasal/efeitos dos fármacos , Nitrito de Sódio/efeitos adversos , Administração por Inalação , Animais , Animais Endogâmicos , Anti-Hipertensivos/administração & dosagem , Anti-Hipertensivos/uso terapêutico , Cães , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Drogas em Investigação/administração & dosagem , Drogas em Investigação/uso terapêutico , Feminino , Hipertensão Pulmonar/tratamento farmacológico , Hipotensão/sangue , Hipotensão/induzido quimicamente , Hipotensão/metabolismo , Hipotensão/patologia , Infusões Intravenosas , Masculino , Metemoglobinemia/sangue , Metemoglobinemia/induzido quimicamente , Metemoglobinemia/metabolismo , Metemoglobinemia/patologia , Cavidade Nasal/imunologia , Cavidade Nasal/metabolismo , Cavidade Nasal/patologia , Mucosa Nasal/imunologia , Mucosa Nasal/metabolismo , Mucosa Nasal/patologia , Nível de Efeito Adverso não Observado , Ratos Sprague-Dawley , Medição de Risco , Nitrito de Sódio/administração & dosagem , Nitrito de Sódio/uso terapêutico , Especificidade da Espécie , Testes de Toxicidade Crônica
3.
J Cardiovasc Pharmacol ; 57(1): 79-85, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20980921

RESUMO

HBI-3000 (sulcardine sulfate) has been shown to suppress various ventricular arrhythmias in animal models. The electrophysiological properties of HBI-3000 were investigated using standard microelectrode and patch-clamp techniques in single human ventricular myocytes. HBI-3000 led to concentration-dependent suppression of dofetilide-induced early afterdepolarizations in single nonfailing human ventricular myocytes and early afterdepolarizations seen in failing ventricular myocytes. The concentration-dependent prolongation of action potential duration (APD) by HBI-3000 was bell shaped with maximum response occurring around 10 µM. Interestingly, HBI-3000 at the concentration of 10 µM modestly prolonged the APD at all 3 basic cycle lengths. The slope of APD-cycle length curve of HBI-3000 was only slightly steeper than that of control (88.8 ± 7.7 ms/s vs. 78.9 ± 5.2 ms/s in control, n = 8, P > 0.05). HBI-3000 only showed a minimal use-dependent prolongation of the APD in human ventricular myocytes. HBI-3000 inhibited fast sodium current (INa-F), late sodium channel (INa-L), L-type calcium current (ICa-L), and rapidly activating delayed rectifier K current (IKr) in single human ventricular myocytes. The estimated half-maximal inhibitory concentration values of INa-F, INa-L, ICa-L, and IKr were 48.3 ± 3.8, 16.5 ± 1.4, 32.2 ± 2.9, and 22.7 ± 2.5 µM, respectively. The ion channel profile and electrophysiological properties of HBI-3000 are similar to those of ranolazine and chronic amiodarone (reduced INa-F, INa-L, ICa-L, and IKr). HBI-3000 may be a promising antiarrhythmic agent with low proarrhythmic risk.


Assuntos
Antiarrítmicos/farmacologia , Fenômenos Eletrofisiológicos , Ventrículos do Coração/metabolismo , Miócitos Cardíacos/fisiologia , Acetanilidas , Potenciais de Ação/efeitos dos fármacos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Ventrículos do Coração/fisiopatologia , Humanos , Células Musculares/metabolismo , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp/métodos , Fenetilaminas , Piperazinas , Ranolazina , Sódio/metabolismo , Sódio/farmacologia , Canais de Sódio/farmacologia , Canais de Sódio/fisiologia , Sulfonamidas , Ésteres do Ácido Sulfúrico
4.
Mol Cancer Ther ; 9(2): 400-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20124448

RESUMO

AMG 102 is a fully human monoclonal antibody that selectively targets and neutralizes hepatocyte growth factor/scatter factor (HGF/SF). A detailed biochemical and functional characterization of AMG 102 was done to support its clinical development for the treatment of cancers dependent on signaling through the HGF/SF:c-Met pathway. In competitive equilibrium binding experiments, AMG 102 bound to human and cynomolgus monkey HGF with affinities of approximately 19 pmol/L and 41 pmol/L, respectively. However, AMG 102 did not detect mouse or rabbit HGF on immunoblots. Immunoprecipitation experiments showed that AMG 102 preferentially bound to the mature, active form of HGF, and incubation of AMG 102/HGF complexes with kallikrein protease indicated that AMG 102 had no apparent effect on proteolytic processing of the inactive HGF precursor. AMG 102 inhibited human and cynomolgus monkey HGF-induced c-Met autophosphorylation in PC3 cells with IC(50) values of 0.12 nmol/L and 0.24 nmol/L, respectively. AMG 102 also inhibited cynomolgus monkey HGF-induced migration of human MDA-MB-435 cells but not rat HGF-induced migration of mouse 4T1 cells. Epitope-mapping studies of recombinant HGF molecules comprising human/mouse chimeras and human-to-mouse amino acid substitutions showed that amino acid residues near the NH(2)-terminus of the beta-chain are critical for AMG 102 binding. Bound AMG 102 protected one trypsin protease cleavage site near the NH(2)-terminus of the beta-chain of human HGF, further substantiating the importance of this region for AMG 102 binding. Currently, AMG 102 is in phase II clinical trials in a variety of solid tumor indications. Mol Cancer Ther; 9(2); 400-9.


Assuntos
Anticorpos Monoclonais/química , Fator de Crescimento de Hepatócito/química , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Mapeamento de Epitopos , Humanos , Immunoblotting , Concentração Inibidora 50 , Macaca fascicularis , Camundongos , Biblioteca de Peptídeos , Fosforilação , Primatas , Coelhos , Proteínas Recombinantes/química
5.
Assay Drug Dev Technol ; 8(2): 228-37, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20035617

RESUMO

Flow cytometry (FCM) is an important technology with a broad spectrum of applications ranging from basic research to clinical diagnostics. In a typical FCM experiment, thousands of cells are queried with respect to size, shape, and abundance of multiple cell surface antigens. Recent advances in FCM techniques and instrumentation have enabled researchers to raise the throughput of experimentation dramatically. However, data analysis has remained a time-consuming activity requiring significant manual intervention for gating as well as for overall data reduction and interpretation. Presented in this article is a novel, algorithmically flexible, internally developed, software framework for the analysis of plate-based FCM data for high-throughput screening (HTS). Utilizing a post-treatment pooling strategy, >87,000 individual wells representing over 240,000 compounds were automatically gated, percent of control (POC) calculated, results assembled, deconvolved, and sorted, allowing researchers to visually assess wells of interest in minutes.


Assuntos
Avaliação Pré-Clínica de Medicamentos/estatística & dados numéricos , Citometria de Fluxo/estatística & dados numéricos , Software , Algoritmos , Linhagem Celular Tumoral , Interpretação Estatística de Dados , Avaliação Pré-Clínica de Medicamentos/instrumentação , Avaliação Pré-Clínica de Medicamentos/métodos , Citometria de Fluxo/instrumentação , Citometria de Fluxo/métodos , Humanos , Bibliotecas de Moléculas Pequenas
6.
Comb Chem High Throughput Screen ; 12(9): 849-59, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19929790

RESUMO

As technologies for high throughput and high content screening continue to evolve, new platforms for quantitative cellular imaging will play an increasingly important role in identifying and profiling lead compounds. To gain insight into the effects of a compound on cell morphology or intracellular events, it is necessary to have quality images and the ability to enumerate thousands of data points for statistical relevance. Imaging flow cytometry combines many of the features of flow cytometry, microscopy and imaging as well as a number of unique characteristics. The result is an instrument capable of highly quantitative analysis of cellular behaviors such as receptor internalization, phagocytosis, cell-cell communication, apoptosis and nuclear translocation. This promising new technology and unique type of flow cytometry provides enhanced capabilities for highly multiplexed assays. Here, we review the capabilities of the ImageStream imaging cytometer and discuss several applications relevant to compound screening and profiling.


Assuntos
Descoberta de Drogas/métodos , Citometria de Fluxo/métodos , Ensaios de Triagem em Larga Escala/métodos , Células Sanguíneas/citologia , Células Sanguíneas/efeitos dos fármacos , Descoberta de Drogas/instrumentação , Citometria de Fluxo/instrumentação , Ensaios de Triagem em Larga Escala/instrumentação , Humanos , Preparações Farmacêuticas , Software
7.
J Pharmacol Exp Ther ; 329(1): 102-11, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19129372

RESUMO

Antagonizing the glucagon signaling pathway represents an attractive therapeutic approach for reducing excess hepatic glucose production in patients with type 2 diabetes. Despite extensive efforts, there is currently no human therapeutic that directly inhibits the glucagon/glucagon receptor pathway. We undertook a novel approach by generating high-affinity human monoclonal antibodies (mAbs) to the human glucagon receptor (GCGR) that display potent antagonistic activity in vitro and in vivo. A single injection of a lead antibody, mAb B, at 3 mg/kg, normalized blood glucose levels in ob/ob mice for 8 days. In addition, a single injection of mAb B dose-dependently lowered fasting blood glucose levels without inducing hypoglycemia and improved glucose tolerance in normal C57BL/6 mice. In normal cynomolgus monkeys, a single injection improved glucose tolerance while increasing glucagon and active glucagon-like peptide-1 levels. Thus, the anti-GCGR mAb could represent an effective new therapeutic for the treatment of type 2 diabetes.


Assuntos
Anticorpos Bloqueadores/farmacologia , Anticorpos Monoclonais/farmacologia , Glucose/metabolismo , Homeostase/efeitos dos fármacos , Receptores de Glucagon/antagonistas & inibidores , Animais , Glicemia/metabolismo , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Endocitose/efeitos dos fármacos , Citometria de Fluxo , Teste de Tolerância a Glucose , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Cinética , Ligantes , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos
8.
Clin Cancer Res ; 14(1): 309-17, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18172283

RESUMO

PURPOSE: Bendamustine has shown clinical activity in patients with disease refractory to conventional alkylator chemotherapy. The purpose of this study was to characterize the mechanisms of action of bendamustine and to compare it with structurally related compounds. EXPERIMENTAL DESIGN: Bendamustine was profiled in the National Cancer Institute in vitro antitumor screen. Microarray-based gene expression profiling, real-time PCR, immunoblot, cell cycle, and functional DNA damage repair analyses were used to characterize response to bendamustine and compare it with chlorambucil and phosphoramide mustard. RESULTS: Bendamustine displays a distinct pattern of activity unrelated to other DNA-alkylating agents. Its mechanisms of action include activation of DNA-damage stress response and apoptosis, inhibition of mitotic checkpoints, and induction of mitotic catastrophe. In addition, unlike other alkylators, bendamustine activates a base excision DNA repair pathway rather than an alkyltransferase DNA repair mechanism. CONCLUSION: These results suggest that bendamustine possesses mechanistic features that differentiate it from other alkylating agents and may contribute to its distinct clinical efficacy profile.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Ciclo Celular/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Compostos de Mostarda Nitrogenada/farmacologia , Apoptose/efeitos dos fármacos , Cloridrato de Bendamustina , Western Blotting , Linhagem Celular Tumoral , Clorambucila/farmacologia , Ciclofosfamida/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Citometria de Fluxo , Humanos
9.
Arch Pharm (Weinheim) ; 340(10): 511-6, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17849446

RESUMO

Non-steroidal anti-inflammatory drugs have been shown to inhibit carcinogenesis in colon cancer, and to induce apoptosis in a variety of tumor cell lines. Some anti-tumor effects are thought to be related to their cyclooxygenase-2-inhibitory activity, but recent studies have shown that non-steroidal anti-inflammatory drugs exert their anti-tumor effect via cyclooxygenase-2-independent mechanism. SDX-308 (CEP-18082) is a non-cyclooxygenase-2-inhibiting indole-pyran analog and is structurally related to SDX-101, an R-enantiomer of etodolac. SDX-308 has a potent anti-myeloma effect and shows synergism in combination with other drugs for the treatment of chronic lymphocytic leukemia. In addition SDX-308 inhibits osteoclast formation and activity and thereby might be an attractive drug for the treatment of diseases with increased osteoclast activity such as osteolytic lesions in multiple myeloma and metastatic carcinomas, as well as osteoporosis. This review covers future application of SDX-308 as an anti-myeloma drug regulating increased osteoclast activity.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Antineoplásicos/uso terapêutico , Etodolac/química , Etodolac/uso terapêutico , Neoplasias Hematológicas/tratamento farmacológico , Compostos Heterocíclicos com 3 Anéis/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , NF-kappa B/antagonistas & inibidores , Anti-Inflamatórios não Esteroides/farmacologia , Antineoplásicos/farmacologia , Etodolac/farmacologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Mieloma Múltiplo/patologia , NF-kappa B/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/fisiologia , Estereoisomerismo
10.
Invest New Drugs ; 25(4): 297-303, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17440681

RESUMO

SDX-308 and SDX-309 are potent indole-pyran analogues of SDX-101 (R-etodolac) which has anti-tumour activity unrelated to cyclooxygenase-2 inhibition. Their cytotoxic activity was further studied herein using a well-characterized human tumour cell-line panel containing ten cell lines, as well as in 58 primary tumour cell samples from a variety of diagnoses. The indole-pyran analogues of SDX-101 were in general considerably more active in both cancer cell lines and primary tumour samples. Low cross-reactivity with standard agents was observed, indicating a unique mechanism of action. No apparent influence on efficacy was observed via classical mechanisms of multidrug-resistance. SDX-101 and SDX-309 showed higher relative activity in haematological compared to solid tumour samples, while SDX-308 had pronounced solid-tumour activity. High SDX-308 cytotoxic efficacy was observed in non-small cell lung cancer, renal cancer and ovarian cancer samples, and also in chronic lymphocytic leukaemia. In conclusion, the indole-pyran analogues showed a favourable pharmacological profile and represent a potentially important new class of drugs for cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Etodolac/análogos & derivados , Etodolac/farmacologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos
11.
Blood ; 109(5): 2130-8, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17095620

RESUMO

Multiple myeloma is characterized by increased osteoclast activity that results in bone destruction and lytic lesions. With the prolonged overall patient survival achieved by new treatment modalities, additional drugs are required to inhibit bone destruction. We focused on a novel and more potent structural analog of the nonsteroidal anti-inflammatory drug etodolac, known as SDX-308, and its effects on osteoclastogenesis and multiple myeloma cells. SDX-101 is another structural analog of etodolac that is already used in clinical trials for the treatment of B-cell chronic lymphocytic leukemia (B-CLL). Compared with SDX-101, a 10-fold lower concentration of SDX-308 induced potent (60%-80%) inhibition of osteoclast formation, and a 10- to 100-fold lower concentration inhibited multiple myeloma cell proliferation. Bone resorption was completely inhibited by SDX-308, as determined in dentin-based bone resorption assays. SDX-308 decreased constitutive and RANKL-stimulated NF-kappaB activation and osteoclast formation in an osteoclast cellular model, RAW 264.7. SDX-308 effectively suppressed TNF-alpha-induced IKK-gamma and IkappaB-alpha phosphorylation and degradation and subsequent NF-kappaB activation in human multiple myeloma cells. These results indicate that SDX-308 effectively inhibits multiple myeloma cell proliferation and osteoclast activity, potentially by controlling NF-kappaB activation signaling. We propose that SDX-308 is a promising therapeutic candidate to inhibit multiple myeloma growth and osteoclast activity and that it should receive attention for further study.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Diferenciação Celular/efeitos dos fármacos , Compostos Heterocíclicos com 3 Anéis/farmacologia , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , NF-kappa B/antagonistas & inibidores , Osteoclastos/efeitos dos fármacos , Animais , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/toxicidade , Reabsorção Óssea/metabolismo , Proliferação de Células/efeitos dos fármacos , Compostos Heterocíclicos com 3 Anéis/química , Compostos Heterocíclicos com 3 Anéis/toxicidade , Humanos , Camundongos , NF-kappa B/metabolismo , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoclastos/citologia , Osteoclastos/metabolismo , Ligante RANK/farmacologia , Transdução de Sinais , Células Tumorais Cultivadas
12.
Cancer Chemother Pharmacol ; 60(4): 545-53, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17186240

RESUMO

OBJECTIVE: SDX-101 is the non-cyclooxygenase 2-inhibiting R-enantiomer of the non-steroid anti-inflammatory drug etodolac, and has anti-tumour activity in chronic lymphocytic leukaemia (CLL). SDX-308 and SDX-309 are more potent, structurally related indole-pyran analogues of SDX-101. The current study was performed to investigate and quantify the cytotoxic potentiating effects resulting from a combination of either SDX-101, SDX-308 or SDX-309 with standard cytotoxic agents used in the CLL treatment today. METHODS: The lymphoma cell line U937-gtb was used, together with primary tumour cells isolated from seven CLL patients. Combinations between chlorambucil and each one of the agents etodolac, SDX-101, SDX-308 and SDX-309 were studied. In addition, SDX-309 was combined with fludarabine, doxorubicin or vincristine. Both simultaneous and sequential exposures were explored using the median-effect method. RESULTS: Most combinations were additive, which could be of clinical benefit since SDX-101 has been shown to be well tolerated. At the 70% effect level, synergy was observed between SDX-308 and chlorambucil in U937-gtb cells and in two-third of the CLL samples. Since chlorambucil is the most important drug in CLL therapy today and SDX-308 is presently targeted as the lead clinical candidate, this combination would be interesting for further studies. Vincristine and SDX-309 were synergistic in two-fourth of CLL samples. CONCLUSIONS: To conclude, the non-COX-inhibiting etodolac-derivatives SDX-101, SDX-308 and SDX-309 are potential candidates for combination treatment of CLL. Especially, SDX-308 in combination with chlorambucil warrants further evaluation.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Etodolac/farmacologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Clorambucila/administração & dosagem , Doxorrubicina/administração & dosagem , Sinergismo Farmacológico , Feminino , Humanos , Linfoma/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Vincristina/administração & dosagem
13.
Br J Haematol ; 134(1): 37-44, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16803565

RESUMO

Glucocorticoids have been widely used in the treatment of multiple myeloma (MM) both as single agents and in combination with other drugs. However, primary or acquired glucocorticoid resistance occurs in most cases. It was recently reported that R-etodolac induced in vitro cytotoxicity in MM cell lines and in primary MM cells, as well as synergistically enhanced dexamethasone (Dex)-induced apoptosis in Dex-sensitive MM.1S cells. This study examined the in vitro and in vivo effects of combination treatment with R-etodolac and Dex on Dex-resistant OPM1 cells. Treatment with R-etodolac and Dex was found to enhance cytotoxicity, inhibit nuclear factor kappaB activity via upregulation of IkappaBalpha, as well as enhance Dex-induced caspase activation and poly (ADP)-ribose polymerase cleavage in OPM1 cells. R-etodolac also enhanced Dex cytotoxicity in patient MM cells that were resistant to glucocorticoids. The in vivo anti-tumour effect of this combination on MM cells was evaluated by using severe combined immunodeficient mice engrafted with OPM1. Treatment with R-etodolac or Dex alone did not induce a significant reduction of tumour volume; in contrast, combination treatment with R-etodolac and Dex induced significant synergistic inhibition of tumour growth. These data indicate that R-etodolac overcomes resistance to Dex in glucocorticoid-resistant MM cells, providing the framework for clinical trials of R-etodolac combined with Dex, to improve patient outcome in MM.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Dexametasona/uso terapêutico , Etodolac/uso terapêutico , Glucocorticoides/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Linhagem Celular Tumoral , Sinergismo Farmacológico , Quimioterapia Combinada , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Proteínas I-kappa B/metabolismo , Immunoblotting , Camundongos , Camundongos SCID , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Poli(ADP-Ribose) Polimerases/metabolismo , Transplante Heterólogo
14.
J Immunol ; 176(7): 4419-30, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16547280

RESUMO

In this study, we describe the identification and in vitro functional activity of a novel multiple domain complement regulatory protein discovered based on its homology to short consensus repeat (SCR)-containing proteins of the regulators of complement activation (RCA) gene family. The rat cDNA encodes a predicted 388-kDa protein consisting of 14 N-terminal CUB domains that are separated from each other by a SCR followed by 15 tandem SCR domains, a transmembrane domain, and a short cytoplasmic tail. This protein is the homolog of the human protein of unknown function called the CUB and sushi multiple domains 1 (CSMD1) protein. A cloning strategy that incorporates the two C-terminal CUB-SCR domains and 12 of the tandem SCR repeats was used to produce a soluble rat CSMD1 protein. This protein blocked classical complement pathway activation in a comparable fashion with rat Crry but did not block alternative pathway activation. Analysis of CSMD1 mRNA expression by in situ hybridization and immunolabeling of neurons indicates that the primary sites of synthesis are the developing CNS and epithelial tissues. Of particular significance is the enrichment of CSMD1 in the nerve growth cone, the amoeboid-leading edge of the growing neuron. These results suggest that CSMD1 may be an important regulator of complement activation and inflammation in the developing CNS, and that it may also play a role in the context of growth cone function.


Assuntos
Sistema Nervoso Central/metabolismo , Epitélio/metabolismo , Proteínas de Membrana/metabolismo , Envelhecimento/fisiologia , Animais , Linhagem Celular , Sistema Nervoso Central/citologia , Clonagem Molecular , Via Clássica do Complemento , Eritrócitos/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica , Cones de Crescimento/metabolismo , Hemólise/efeitos dos fármacos , Humanos , Hibridização In Situ , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Proteínas de Membrana/farmacologia , Especificidade de Órgãos , RNA Mensageiro/análise , RNA Mensageiro/genética , Ratos , Ovinos , Solubilidade
15.
Mol Cancer Ther ; 5(2): 337-46, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16505107

RESUMO

L-alanosine (SDX-102) exerts its cytotoxicity through inhibition of de novo purine biosynthesis, an effect potentiated by methylthioadenosine phosphorylase (MTAP) deficiency. The relevance of circadian dosing time was investigated for chronotherapeutic optimization of SDX-102. Toxicity was assessed in healthy mice following single (1,150, 1,650, or 1,850 mg/kg/d) or multiple doses (250 or 270 mg/kg/d). Efficacy was tested in mice with P388 leukemia receiving multiple doses (225 or 250 mg/kg/d). SDX-102 was administered at six circadian times 4 hours apart in mice synchronized with 12 hours of light alternating with 12 hours of darkness. MTAP expression was determined in liver, bone marrow, small intestinal mucosa, and P388 cells. Dosing at 19 hours after light onset reduced lethality 5-fold after single administration and 3-fold after multiple doses as compared with worst time [P < 0.001 and P < 0.01, respectively (chi2 test)]. Neutropenia, lymphopenia, and bone marrow hemorrhagic lesions were significantly less in mice dosed at 19 hours after light onset as compared with 7 hours after light onset. SDX-102 at 7 hours after light onset transiently ablated the 24-hour patterns in body temperature and activity. A circadian rhythm characterized small intestinal MTAP expression with a maximum at 6:30 hours after light onset (P = 0.04). A minor survival improvement was found in MTAP-deficient P388 mice receiving SDX-102 at 7 or 23 hours after light onset as compared with other times (P = 0.03, log-rank test). In conclusion, the therapeutic index of SDX-102 was improved by the delivery of SDX-102 in the mid to late activity span. These results support the concept of chronomodulated infusion of SDX-102 in cancer patients.


Assuntos
Antineoplásicos/toxicidade , Cronoterapia , Leucemia P388/tratamento farmacológico , Alanina/administração & dosagem , Alanina/análogos & derivados , Alanina/uso terapêutico , Alanina/toxicidade , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Peso Corporal/efeitos dos fármacos , Medula Óssea/efeitos dos fármacos , Medula Óssea/enzimologia , Escuridão , Expressão Gênica , Hemorragia/induzido quimicamente , Intestinos/efeitos dos fármacos , Intestinos/enzimologia , Intestinos/patologia , Luz , Fígado/efeitos dos fármacos , Fígado/enzimologia , Linfopenia/induzido quimicamente , Masculino , Camundongos , Neutropenia/induzido quimicamente , Purina-Núcleosídeo Fosforilase/análise , Purina-Núcleosídeo Fosforilase/genética , Purina-Núcleosídeo Fosforilase/metabolismo , Resultado do Tratamento
16.
Cancer Res ; 66(3): 1721-9, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16452232

RESUMO

c-Met is a well-characterized receptor tyrosine kinase for hepatocyte growth factor (HGF). Compelling evidence from studies in human tumors and both cellular and animal tumor models indicates that signaling through the HGF/c-Met pathway mediates a plethora of normal cellular activities, including proliferation, survival, migration, and invasion, that are at the root of cancer cell dysregulation, tumorigenesis, and tumor metastasis. Inhibiting HGF-mediated signaling may provide a novel therapeutic approach for treating patients with a broad spectrum of human tumors. Toward this goal, we generated and characterized five different fully human monoclonal antibodies that bound to and neutralized human HGF. Antibodies with subnanomolar affinities for HGF blocked binding of human HGF to c-Met and inhibited HGF-mediated c-Met phosphorylation, cell proliferation, survival, and invasion. Using a series of human-mouse chimeric HGF proteins, we showed that the neutralizing antibodies bind to a unique epitope in the beta-chain of human HGF. Importantly, these antibodies inhibited HGF-dependent autocrine-driven tumor growth and caused significant regression of established U-87 MG tumor xenografts. Treatment with anti-HGF antibody rapidly inhibited tumor cell proliferation and significantly increased the proportion of apoptotic U-87 MG tumor cells in vivo. These results suggest that an antibody to an epitope in the beta-chain of HGF has potential as a novel therapeutic agent for treating patients with HGF-dependent tumors.


Assuntos
Anticorpos Monoclonais/farmacologia , Glioblastoma/terapia , Fator de Crescimento de Hepatócito/imunologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/imunologia , Linhagem Celular Tumoral , Epitopos/imunologia , Feminino , Glioblastoma/imunologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Imunoglobulina G/biossíntese , Imunoglobulina G/química , Imunoglobulina G/imunologia , Camundongos , Camundongos Nus , Fosforilação , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Blood ; 106(2): 706-12, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15802527

RESUMO

In this study we report that R-etodolac (SDX-101), at clinically relevant concentrations, induces potent cytotoxicity in drug-sensitive multiple myeloma (MM) cell lines, as well as in dexamethasone (MM.1R)-, doxorubicin (Dox40/RPMI8226)-, and bortezomib (DHL4)-resistant cell lines. Immunoblot analysis demonstrates that R-etodolac induces apoptosis characterized by caspase-8, -9, and -3 and PARP (poly-ADP [adenosine diphosphate]-ribose polymerase) cleavage and down-regulation of cyclin D1 expression. Subcytotoxic doses of R-etodolac up-regulate myeloid cell leukemia-1 proapoptotic variant (Mcl-1S), while enhancing dexamethasone (Dex)-induced caspase activation and apoptosis. The combination of R-etodolac with Dex results in a highly synergistic cytotoxic effect. R-etodolac also induces apoptosis against primary cells isolated from patients with MM refractory to chemotherapy. Although interleukin 6 (IL-6) and insulin-like growth factor-1 (IGF-1) abrogate Dex-induced MM cell cytotoxicity, neither IL-6 nor IGF-1 protects against R-etodolac-induced cytotoxicity in MM cells. R-etodolac also inhibits viability of MM cells adherent to bone marrow stromal cells (BMSCs), thereby overcoming a mechanism of drug resistance commonly observed with other conventional chemotherapeutic agents. Our data, therefore, indicate that R-etodolac circumvents drug resistance in MM cells at clinically relevant concentrations, targets Mcl-1, and can be synergistically combined with Dex.


Assuntos
Antineoplásicos/farmacologia , Dexametasona/administração & dosagem , Etodolac/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/patologia , Caspases/metabolismo , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ciclina D1/metabolismo , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Etodolac/administração & dosagem , Etodolac/química , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Interleucina-6/farmacologia , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas de Neoplasias/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Estereoisomerismo
18.
Artigo em Inglês | MEDLINE | ID: mdl-15063341

RESUMO

A sensitive method was developed for quantitation of the cytotoxic antibiotic l-alanosine in human plasma. Alanosine was extracted from plasma by anion-exchange solid phase extraction, derivatized with dansyl chloride and analyzed by liquid chromatography-tandem mass spectrometry using atmospheric pressure chemical ionization in negative mode. Dansylation led to 50-fold improvement of method sensitivity over non-dansylated alanosine with a resulting 20 ng/ml limit of alanosine quantitation in plasma being achieved. The method was validated and applied for clinical studies of alanosine administered to cancer patients.


Assuntos
Alanina/sangue , Antibióticos Antineoplásicos/sangue , Antineoplásicos/sangue , Cromatografia por Troca Iônica/métodos , Espectrometria de Massas/métodos , Alanina/análogos & derivados , Sensibilidade e Especificidade
19.
Clin Cancer Res ; 9(10 Pt 2): 3982S-90S, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-14506197

RESUMO

PURPOSE: Epratuzumab is a novel humanized antihuman CD22 IgG1 antibody that has recently shown promising clinical activity, both as a single agent and in combination with rituximab, in patients with non-Hodgkin's lymphomas (NHL). In an attempt to better understand the mode of action of epratuzumab, the antibody was tested in vitro in a variety of cell-based assays similar to those used to evaluate the biological activity of other therapeutic monoclonal antibodies, including rituximab. In this report, we present epratuzumab activities as they relate to binding, signaling, and internalization of the receptor CD22. METHODS: Chinese hamster ovary-expressed CD22 extracellular domain was used to measure epratuzumab affinity on Biacore. CD22 receptor density and internalization rate were measured indirectly using a monovalently labeled, noncompeting (with epratuzumab) anti-CD22 antibody on Burkitt lymphoma cell lines, primary B cells derived from fresh tonsils, and B cells separated from peripheral blood samples obtained from patients with chronic lymphocytic leukemia or healthy volunteers. Epratuzumab-induced CD22 phosphorylation was measured by immunoprecipitation/Western blot and compared with that induced by anti-IgM stimulation. RESULTS: Epratuzumab binds to CD22-extracellular domain, with an affinity of K(D) = 0.7 nM. Binding of epratuzumab to B cell lines, or primary B cells from healthy individuals and patients with NHL, results in rapid internalization of the CD22/antibody complex. Internalization appears to be faster at early time points in cell lines than in primary B cells and NHL patient-derived B cells, but the maximum internalization reached is comparable for all B cell populations after several hours of treatment and appears to reach saturation at antibody concentrations of 1-5 micro g/ml. Finally, epratuzumab binding results in modest but significant CD22 phosphorylation. CONCLUSIONS: Epratuzumab represents an excellent anti-CD22 ligating agent, highly efficacious in inducing CD22 internalization, and can induce phosphorylation. Although we cannot unequivocally demonstrate here that epratuzumab-induced internalization and signaling of CD22 directly contribute to its therapeutic efficacy, these properties are the fundamental characteristics of the target CD22 and its interaction with epratuzumab. Similar results were observed when epratuzumab was tested in vitro on Burkitt B cell lines as well as on primary normal B cells and neoplastic B cells separated from fresh peripheral blood samples from patients with chronic lymphocytic leukemia.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígenos CD/biossíntese , Antígenos de Diferenciação de Linfócitos B/biossíntese , Moléculas de Adesão Celular , Lectinas/biossíntese , Animais , Anticorpos Monoclonais Humanizados , Anticorpos Monoclonais Murinos , Antígenos CD/genética , Antígenos CD19/biossíntese , Antígenos CD20/biossíntese , Antígenos de Diferenciação de Linfócitos B/genética , Western Blotting , Células CHO , Linhagem Celular , Linhagem Celular Tumoral , Clonagem Molecular , Cricetinae , Humanos , Imunoglobulina M/química , Técnicas In Vitro , Cinética , Lectinas/genética , Microscopia Confocal , Fosforilação , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Rituximab , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico , Fatores de Tempo
20.
Proc Natl Acad Sci U S A ; 100(9): 5431-6, 2003 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-12702766

RESUMO

The tumor suppressor p53 is regulated in part by binding to cellular proteins. We used p53 as bait in the yeast two-hybrid system and isolated homeodomain-interacting protein kinase 1 (HIPK1) as a p53-binding protein. Deletion analysis showed that amino acids 100-370 of p53 and amino acids 885-1093 of HIPK1 were sufficient for HIPK1-p53 interaction. HIPK1 was capable of autophosphorylation and specific serine phosphorylation of p53. The HIPK1 gene was highly expressed in human breast cancer cell lines and oncogenically transformed mouse embryonic fibroblasts. HIPK1 was localized to human chromosome band 1p13, a site frequently altered in cancers. Gene-targeted HIPK1-/- mice were grossly normal but oncogenically transformed HIPK1 -/- mouse embryonic fibroblasts exhibited reduced transcription of Mdm2 and were more susceptible than transformed HIPK1+/+ cells to apoptosis induced by DNA damage. Carcinogen-treated HIPK1 -/- mice developed fewer and smaller skin tumors than HIPK1+/+ mice. HIPK1 may thus play a role in tumorigenesis, perhaps by means of the regulation of p53 and/or Mdm2.


Assuntos
Proteínas de Transporte/genética , Marcação de Genes , Proteínas Quinases/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Northern Blotting , Proteínas de Transporte/metabolismo , Humanos , Camundongos , Camundongos Knockout , Ligação Proteica , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...