Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Transl Med ; 6(240): 240ra75, 2014 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-24920661

RESUMO

Many solid tumors contain an overabundance of phospholipid ethers relative to normal cells. Capitalizing on this difference, we created cancer-targeted alkylphosphocholine (APC) analogs through structure-activity analyses. Depending on the iodine isotope used, radioiodinated APC analog CLR1404 was used as either a positron emission tomography (PET) imaging ((124)I) or molecular radiotherapeutic ((131)I) agent. CLR1404 analogs displayed prolonged tumor-selective retention in 55 in vivo rodent and human cancer and cancer stem cell models. (131)I-CLR1404 also displayed efficacy (tumor growth suppression and survival extension) in a wide range of human tumor xenograft models. Human PET/CT (computed tomography) and SPECT (single-photon emission computed tomography)/CT imaging in advanced-cancer patients with (124)I-CLR1404 or (131)I-CLR1404, respectively, demonstrated selective uptake and prolonged retention in both primary and metastatic malignant tumors. Combined application of these chemically identical APC-based radioisosteres will enable personalized dual modality cancer therapy of using molecular (124)I-CLR1404 tumor imaging for planning (131)I-CLR1404 therapy.


Assuntos
Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Fosforilcolina/uso terapêutico , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Fosforilcolina/análogos & derivados , Tomografia por Emissão de Pósitrons/métodos , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Tomografia Computadorizada por Raios X/métodos , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Cancer Res ; 72(9): 2383-93, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22406622

RESUMO

Many cancer drugs impact cancer cell redox regulatory mechanisms and disrupt redox homeostasis. Pharmacodynamic biomarkers that measure therapeutic efficacy or toxicity could improve patient management. Using immunoblot analyses and mass spectrometry, we identified that serpins A1 and A3 were S-glutathionylated in a dose- and time-dependent manner following treatment of mice with drugs that alter reactive oxygen or nitrogen species. Tandem mass spectrometry analyses identified Cys(256) of serpin A1 and Cys(263) of serpin A3 as the S-glutathionylated residues. In human plasma from cancer patients, there were higher levels of unmodified serpin A1 and A3, but following treatments with redox active drugs, relative S-glutathionylation of these serpins was higher in plasma from normal individuals. There is potential for S-glutathionylated serpins A1 and A3 to act as pharmacodynamic biomarkers for evaluation of patient response to drugs that target redox pathways.


Assuntos
Biomarcadores Tumorais/sangue , Glutationa/análogos & derivados , Neoplasias/sangue , Neoplasias/tratamento farmacológico , alfa 1-Antiquimotripsina/sangue , alfa 1-Antitripsina/sangue , Animais , Cisplatino/farmacologia , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Glutationa/sangue , Dissulfeto de Glutationa/farmacologia , Humanos , Imunoprecipitação , Camundongos , Oxirredução/efeitos dos fármacos , Serpinas/sangue , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem
3.
Free Radic Biol Med ; 52(9): 1560-8, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22343421

RESUMO

The oxidized glutathione mimetic NOV-002 is a unique anti-tumor agent that not only has the ability to inhibit tumor cell proliferation, survival, and invasion, but in some settings can also ameliorate cytotoxic chemotherapy-induced hematopoietic and immune suppression. However, the mechanisms by which NOV-002 protects the hematopoietic and immune systems against the cytotoxic effects of chemotherapy are not known. Therefore, in this study we investigated the mechanisms of action of NOV-002 using a mouse model in which hematopoietic and immune suppression was induced by cyclophosphamide (CTX) treatment. We found that NOV-002 treatment in a clinically comparable dose regimen attenuated CTX-induced reduction in bone marrow hematopoietic stem and progenitor cells (HSPCs) and reversed the immunosuppressive activity of myeloid-derived suppressor cells (MDSCs), which led to a significant improvement in hematopoietic and immune functions. These effects of NOV-002 may be attributable to its ability to modulate cellular redox. This suggestion is supported by the finding that NOV-002 treatment upregulated the expression of superoxide dismutase 3 and glutathione peroxidase 2 in HSPCs, inhibited CTX-induced increases in reactive oxygen species production in HSPCs and MDSCs, and attenuated CTX-induced reduction of the ratio of reduced glutathione to oxidized glutathione in splenocytes. These findings provide a better understanding of the mechanisms whereby NOV-002 modulates chemotherapy-induced myelosuppression and immune dysfunction and a stronger rationale for clinical utilization of NOV-002 to reduce chemotherapy-induced hematopoietic and immune suppression.


Assuntos
Cisplatino/farmacologia , Ciclofosfamida/antagonistas & inibidores , Dissulfeto de Glutationa/farmacologia , Hematopoese/efeitos dos fármacos , Sistema Imunitário/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Animais , Sequência de Bases , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Ciclofosfamida/efeitos adversos , Primers do DNA , Combinação de Medicamentos , Citometria de Fluxo , Glutationa Peroxidase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Superóxido Dismutase/metabolismo
4.
J Biol Chem ; 284(1): 436-445, 2009 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-18990698

RESUMO

Glutathione S-transferase Pi (GSTpi) is a marker protein in many cancers and high levels are linked to drug resistance, even when the selecting drug is not a substrate. S-Glutathionylation of proteins is critical to cellular stress response, but characteristics of the forward reaction are not known. Our results show that GSTpi potentiates S-glutathionylation reactions following oxidative and nitrosative stress in vitro and in vivo. Mutational analysis indicated that the catalytic activity of GST is required. GSTpi is itself redox-regulated. S-Glutathionylation on Cys47 and Cys101 autoregulates GSTpi, breaks ligand binding interactions with c-Jun NH2-terminal kinase (JNK), and causes GSTpi multimer formation, all critical to stress response. Catalysis of S-glutathionylation at low pK cysteines in proteins is a novel property for GSTpi and may be a cause for its abundance in tumors and cells resistant to a range of mechanistically unrelated anticancer drugs.


Assuntos
Glutationa S-Transferase pi/metabolismo , Glutationa/metabolismo , Estresse Oxidativo/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Animais , Resistencia a Medicamentos Antineoplásicos/fisiologia , Glutationa/genética , Glutationa S-Transferase pi/genética , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo
5.
Expert Opin Investig Drugs ; 17(7): 1075-83, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18549343

RESUMO

BACKGROUND: Oxidative signaling to modulate redox-sensitive cell functions is a heretofore unexploited approach to developing new drugs for poorly treated oncology indications, where current therapies are often only palliative and accompanied by severe toxicities. OBJECTIVE: Clinical and non-clinical findings with NOV-002 (a mimetic of glutathione disulfide that represents such an approach) are reviewed and evaluated. METHODS: Published data on NOV-002 along with unpublished information from the drug's sponsor were reviewed. Literature analysis also focused on protein S-glutathionylation as a regulatory mechanism, particularly in relation to cell signaling, proliferation and cytoskeletal architecture. RESULTS/CONCLUSION: NOV-002 is a mechanistically novel agent with potential for ameliorating hematologic toxicity and enhancing efficacy when used in combination with standard chemotherapy to treat cancer patients.


Assuntos
Cisplatino/química , Cisplatino/farmacologia , Dissulfeto de Glutationa/química , Dissulfeto de Glutationa/farmacologia , Mimetismo Molecular , Animais , Cisplatino/efeitos adversos , Cisplatino/metabolismo , Combinação de Medicamentos , Tolerância a Medicamentos , Dissulfeto de Glutationa/efeitos adversos , Dissulfeto de Glutationa/metabolismo , Humanos , Estrutura Molecular
6.
Cancer Res ; 68(8): 2870-7, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18413755

RESUMO

NOV-002 is a novel glutathione disulfide mimetic that when administered in combination with standard chemotherapeutic regimens has resulted in increased efficacy (survival, tumor response) and improved tolerance to chemotherapy (e.g., hematologic recovery) in advanced non-small cell lung cancer patients. We show that NOV-002, which is not cytotoxic as a single agent, generated time- and concentration-dependent oxidative signals at the cell surface (reduction in protein thiols) and intracellularly [altered oxidized glutathione (GSSG) and reduced glutathione levels and ratio; increased reactive oxygen species] in the premyeloid HL-60 cell line and that this was associated with an increase in S-glutathionylation of cell proteins, particularly actin. Commensurate with these effects, NOV-002 activated p38, c-Jun-NH(2)-kinase, and extracellular signal-regulated kinase and caused a dose-dependent increase in phosphorylation of three proteins that have previously been linked with hematopoiesis, AKT, JAK2, and STAT5. The effect of NOV-002 on enzymes involved in glutathione metabolism was evaluated. Relative to oxidized glutathione, NOV-002 was an equivalent substrate for glutathione reductase and was an inhibitor of protein disulfide isomerase, one of the components of the redox-sensitive unfolded protein response pathway. These redox-stimulated cell signaling actions occurred in the context of increased HL-60 cell proliferation after treatment with NOV-002. Overall, the pleiotropic pharmacologic effects of NOV-002 can be attributed to the GSSG component of the drug, and modulation of cellular redox balance is a feature central to the mechanism of action of NOV-002. Such modulation may underlie its clinical actions, including hematologic recovery and immunostimulation in the face of chemosuppression.


Assuntos
Antineoplásicos/farmacologia , Fenômenos Fisiológicos Celulares/efeitos dos fármacos , Cisplatino/farmacologia , Dissulfeto de Glutationa/farmacologia , Glutationa/metabolismo , Carcinoma Pulmonar de Células não Pequenas , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/análogos & derivados , Combinação de Medicamentos , Humanos , Neoplasias Pulmonares , Oxirredução
7.
Pain ; 21(4): 329-337, 1985 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-4000684

RESUMO

Potentiation of the antinociceptive effects of morphine by the tricyclic antidepressants was assayed in awake restrained rats using the tail-flick test. Intrathecally administered amitriptyline, desipramine or sertraline at doses that had no effect themselves (25-30 micrograms) potentiated a subthreshold parenteral dose of morphine (0.5 mg/kg). The morphine potentiating effect of amitriptyline was prevented by prior administration of parachlorophenylalanine (PCPA). This effect of PCPA was not restored by 5-hydroxytryptophan (5-HTP) but was restored when the animals were left for 14 days to replete. The morphine potentiating effects of amitriptyline, desipramine and sertraline were blocked by intrathecal administration of low doses of the serotonin antagonist methysergide and the alpha-adrenergic antagonists yohimbine and phentolamine but not by the beta-adrenergic antagonist propranolol. The results are consistent with the hypothesis that the potentiation of morphine's antinociceptive effect by tricyclic antidepressants depends on activation of both spinopetal serotonin and adrenergic neurons.


Assuntos
Antidepressivos Tricíclicos/farmacologia , Morfina/farmacologia , Nociceptores/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , 1-Naftilamina/análogos & derivados , 1-Naftilamina/farmacologia , Amitriptilina/farmacologia , Animais , Antidepressivos Tricíclicos/antagonistas & inibidores , Desipramina/farmacologia , Sinergismo Farmacológico , Masculino , Norepinefrina/fisiologia , Ratos , Ratos Endogâmicos , Serotonina/fisiologia , Antagonistas da Serotonina/farmacologia , Sertralina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA