Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nanomedicine ; 12(8): 2331-2340, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27335341

RESUMO

Increased levels of soluble amyloid-beta (Aß) oligomers are suspected to underlie Alzheimer's disease (AD) pathophysiology. These oligomers have been shown to form multi-subunit Aß pores in bilayers and induce uncontrolled, neurotoxic, ion flux, particularly calcium ions, across cellular membranes that might underlie cognitive impairment in AD. Small molecule interventions that modulate pore activity could effectively prevent or ameliorate their toxic activity. Here we examined the efficacy of a small molecule, NPT-440-1, on modulating amyloid pore permeability. Co-incubation of B103 rat neuronal cells with NPT-440-1 and Aß1-42 prevented calcium influx. In purified lipid bilayers, we show that a 10-15min preincubation, prior to membrane introduction, was required to prevent conductance. Thioflavin-T and circular dichroism both suggested a reduction in Aß1-42 ß-sheet content during this incubation period. Combined with previous studies on site-specific amino acid substitutions, these results suggest that pharmacological modulation of Aß1-42 could prevent amyloid pore-mediated AD pathogenesis.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Bicamadas Lipídicas , Fragmentos de Peptídeos/metabolismo , Amiloide , Animais , Ratos
2.
Neurobiol Dis ; 74: 144-57, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25173807

RESUMO

Aß accumulation plays a central role in the pathogenesis of Alzheimer's disease (AD). Recent studies suggest that the process of Aß nucleated polymerization is essential for Aß fibril formation, pathology spreading and toxicity. Therefore, targeting this process represents an effective therapeutic strategy to slow or block disease progression. To discover compounds that might interfere with the Aß seeding capacity, toxicity and pathology spreading, we screened a focused library of FDA-approved drugs in vitro using a seeding polymerization assay and identified small molecule inhibitors that specifically interfered with Aß seeding-mediated fibril growth and toxicity. Mitoxantrone, bithionol and hexachlorophene were found to be the strongest inhibitors of fibril growth and protected primary cortical neuronal cultures against Aß-induced toxicity. Next, we assessed the effects of these three inhibitors in vivo in the mThy1-APPtg mouse model of AD (8-month-old mice). We found that mitoxantrone and bithionol, but not hexachlorophene, stabilized diffuse amyloid plaques, reduced the levels of Aß42 oligomers and ameliorated synapse loss, neuronal damage and astrogliosis. Together, our findings suggest that targeting fibril growth and Aß seeding capacity constitutes a viable and effective strategy for protecting against neurodegeneration and disease progression in AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/efeitos dos fármacos , Degeneração Neural/tratamento farmacológico , Degeneração Neural/fisiopatologia , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/efeitos dos fármacos , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/toxicidade , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Bitionol/farmacocinética , Bitionol/farmacologia , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Gliose/tratamento farmacológico , Gliose/patologia , Gliose/fisiopatologia , Hexaclorofeno/farmacocinética , Hexaclorofeno/farmacologia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitoxantrona/farmacocinética , Mitoxantrona/farmacologia , Degeneração Neural/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neurônios/fisiologia , Fármacos Neuroprotetores/farmacocinética , Fragmentos de Peptídeos/toxicidade , Placa Amiloide/tratamento farmacológico , Placa Amiloide/patologia , Placa Amiloide/fisiopatologia , Ratos
3.
J Neuroinflammation ; 12: 236, 2015 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-26683203

RESUMO

BACKGROUND: Deposition of α-synuclein and neuroinflammation are key pathological features of Parkinson's disease (PD). There is no cure for the disease; however, targeting the pathological features might be available to modulate the disease onset and progression. Hypoestoxide (HE) has been demonstrated as a NF-κB modulator, thereby acting as a potential anti-inflammatory and anti-cancer drug. METHODS: In order to assess the effect of HE in a mouse model of PD, mThy1-α-syn transgenic mice received intraperitoneal (IP) injections of either vehicle or HE (5 mg/kg) daily for 4 weeks. RESULTS: Treatment of HE decreased microgliosis, astrogliosis, and pro-inflammatory cytokine gene expression in α-syn transgenic mice. HE administration also prevented the loss of dopaminergic neurons and ameliorated motor behavioral deficits in the α-syn transgenic mice, and α-synuclein pathology was significantly reduced by treatment of HE. In addition, increased levels of nuclear phosphorylated NF-κB in the frontal cortex of α-syn transgenic mice were significantly reduced by HE administration. CONCLUSIONS: These results support the therapeutic potential of HE for PD and other α-synuclein-related diseases.


Assuntos
Modelos Animais de Doenças , Diterpenos/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , alfa-Sinucleína/antagonistas & inibidores , alfa-Sinucleína/biossíntese , Animais , Diterpenos/farmacologia , Feminino , Humanos , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/metabolismo , Camundongos , Camundongos Transgênicos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Neocórtex/efeitos dos fármacos , Neocórtex/metabolismo , Doença de Parkinson/genética
4.
Sci Transl Med ; 15(695): eabq6089, 2023 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-37163617

RESUMO

Alterations in the p38 mitogen-activated protein kinases (MAPKs) play an important role in the pathogenesis of dementia with Lewy bodies (DLB) and Parkinson's disease (PD). Activation of the p38α MAPK isoform and mislocalization of the p38γ MAPK isoform are associated with neuroinflammation and synaptic degeneration in DLB and PD. Therefore, we hypothesized that p38α might be associated with neuronal p38γ distribution and synaptic dysfunction in these diseases. To test this hypothesis, we treated in vitro cellular and in vivo mouse models of DLB/PD with SKF-86002, a compound that attenuates inflammation by inhibiting p38α/ß, and then investigated the effects of this compound on p38γ and neurodegenerative pathology. We found that inhibition of p38α reduced neuroinflammation and ameliorated synaptic, neurodegenerative, and motor behavioral deficits in transgenic mice overexpressing human α-synuclein. Moreover, treatment with SKF-86002 promoted the redistribution of p38γ to synapses and reduced the accumulation of α-synuclein in mice overexpressing human α-synuclein. Supporting the potential value of targeting p38 in DLB/PD, we found that SKF-86002 promoted the redistribution of p38γ in neurons differentiated from iPS cells derived from patients with familial PD (carrying the A53T α-synuclein mutation) and healthy controls. Treatment with SKF-86002 ameliorated α-synuclein-induced neurodegeneration in these neurons only when microglia were pretreated with this compound. However, direct treatment of neurons with SKF-86002 did not affect α-synuclein-induced neurotoxicity, suggesting that SKF-86002 treatment inhibits α-synuclein-induced neurotoxicity mediated by microglia. These findings provide a mechanistic connection between p38α and p38γ as well as a rationale for targeting this pathway in DLB/PD.


Assuntos
Proteína Quinase 14 Ativada por Mitógeno , Doença de Parkinson , Humanos , Camundongos , Animais , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/patologia , alfa-Sinucleína/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Doenças Neuroinflamatórias , Neurônios/metabolismo , Camundongos Transgênicos
5.
Bioconjug Chem ; 23(5): 1003-9, 2012 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-22515422

RESUMO

Dextran-coated superparamagnetic iron oxide nanoparticles (dextran-SPIO conjugates) offer the attractive possibility of enhancing MRI imaging sensitivity so that small or diffuse lesions can be detected. However, systemically injected SPIOs are rapidly removed by macrophages. We engineered embryonic cells (HEK293T) to express major macrophage scavenger receptor (SR) subtypes including SR-AI, MARCO, and endothelial receptor collectin-12. These SRs possess a positively charged collagen-like (CL) domain and they promoted SPIO uptake, while the charge neutral lipoprotein receptor SR-BI did not. In silico modeling indicated a positive net charge on the CL domain and a net negative charge on the cysteine-rich (CR) domain of MARCO and SR-AI. In vitro experiments revealed that CR domain deletion in SR-AI boosted uptake of SPIO 3-fold, while deletion of MARCO's CR domain abolished this uptake. These data suggest that future studies might productively focus on the validation and further exploration of SR charge fields in SPIO recognition.


Assuntos
Meios de Contraste/metabolismo , Dextranos/metabolismo , Macrófagos/metabolismo , Nanopartículas/metabolismo , Receptores Depuradores/metabolismo , Clonagem Molecular , Colágeno Tipo I/metabolismo , Células HEK293 , Humanos , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita , Modelos Moleculares , Nanopartículas/ultraestrutura , Estrutura Terciária de Proteína , Receptores Depuradores/química , Receptores Depuradores/genética
6.
Bioconjug Chem ; 22(8): 1638-44, 2011 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-21786821

RESUMO

Quantum dots (QDs) are attracting intense interest as fluorescence labeling agents for biomedical imaging because biocompatible coatings and relatively nontoxic rare earth metal QDs have emerged as possible options. QD photoemissions are bright, of narrow wavelength range, and very stable. We sought to encapsulate QDs within targeted PEGylated liposomes to reduce their propensity for liver uptake and to amplify the already strong QD emission signal. A novel lipid-QD conjugate initialized a process by which lipids in solution coalesced around the QDs. The liposomal structure was confirmed with size measurements, SEM, and IR spectroscopy. PEGylated QD liposomes injected into a xenograft tumor model largely cleared from the body within 24 h. Residual liver labeling was low. Targeted QD liposomes exhibited robust tumor labeling compared with controls. This study highlights the potential of these near IR emitting QD liposomes for preclinical/clinical applications.


Assuntos
Lipossomos/farmacocinética , Neoplasias Experimentais/tratamento farmacológico , Pontos Quânticos , Animais , Cápsulas , Corantes Fluorescentes , Lipossomos/administração & dosagem , Fígado/metabolismo , Camundongos , Nanopartículas/administração & dosagem , Nanopartículas/uso terapêutico , Polietilenoglicóis , Transplante Heterólogo
7.
Proc Natl Acad Sci U S A ; 105(7): 2313-8, 2008 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-18268346

RESUMO

Screening for novel anticancer drugs in chemical libraries isolated from marine organisms, we identified the lipopeptide somocystinamide A (ScA) as a pluripotent inhibitor of angiogenesis and tumor cell proliferation. The antiproliferative activity was largely attributable to induction of programmed cell death. Sensitivity to ScA was significantly increased among cells expressing caspase 8, whereas siRNA knockdown of caspase 8 increased survival after exposure to ScA. ScA rapidly and efficiently partitioned into liposomes while retaining full antiproliferative activity. Consistent with the induction of apoptosis via the lipid compartment, we noted accumulation and aggregation of ceramide in treated cells and subsequent colocalization with caspase 8. Angiogenic endothelial cells were extremely sensitive to ScA. Picomolar concentrations of ScA disrupted proliferation and endothelial tubule formation in vitro. Systemic treatment of zebrafish or local treatment of the chick chorioallantoic membrane with ScA resulted in dose-dependent inhibition of angiogenesis, whereas topical treatment blocked tumor growth among caspase-8-expressing tumors. Together, the results reveal an unexpected mechanism of action for this unique lipopeptide and suggest future development of this and similar agents as antiangiogenesis and anticancer drugs.


Assuntos
Apoptose/efeitos dos fármacos , Caspase 8/metabolismo , Dissulfetos/farmacologia , Lipoproteínas/farmacologia , Inibidores da Angiogênese/farmacologia , Animais , Animais Geneticamente Modificados , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Galinhas , Dissulfetos/química , Embrião não Mamífero/irrigação sanguínea , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/embriologia , Humanos , Estrutura Molecular , Oceanos e Mares , Fosfolipídeos/metabolismo , Sensibilidade e Especificidade
8.
Nanomedicine ; 6(6): 797-807, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20599526

RESUMO

We report here the in vivo combined-modality imaging of multifunctional drug delivery nanoparticles. These dextran core-based stealth liposomal nanoparticles (nanosomes) contained doxorubicin, iron oxide for magnetic resonance imaging (MRI) contrast, and BODIPY for fluorescence. The particles were long-lived in vivo because of surface decoration with polyethylene glycol and the incorporation of acetylated lipids that were ultraviolet cross-linked for physical stability. We developed a rodent dorsal skinfold window chamber that facilitated both MRI and non-invasive optical imaging of nanoparticle accumulation in the same tumors. Chamber tumors were genetically labeled with DsRed-2, which enabled co-localization of the MR images, the red fluorescence of the tumor, and the blue fluorescence of the nanoparticles. The nanoparticle design and MR imaging developed with the window chamber were then extended to orthotopic pancreatic tumors expressing DsRed-2. The tumors were MR-imaged using iron oxide-dextran liposomes and by fluorescence to demonstrate the deep imaging capability of these nanoparticles.


Assuntos
Doxorrubicina/química , Doxorrubicina/uso terapêutico , Fluorescência , Imageamento por Ressonância Magnética/métodos , Nanopartículas/administração & dosagem , Nanopartículas/química , Animais , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Humanos , Lipossomos/química , Camundongos , Neoplasias/tratamento farmacológico
9.
Beilstein J Org Chem ; 6: 742-7, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20978615

RESUMO

We have developed a synthetic route for the preparation of a hybrid bisubstrate small molecule based on a nucleoside. A prototype compound was designed and docked into the catalytic domain of the AdSS enzyme bridging the region between the magnesium center of the protein to the nucleoside region. The synthesis involves coupling a brominated peptide fragment capable of complexing magnesium to a thiolated nucleoside to obtain the hybrid model compound.

10.
Oncogene ; 22(57): 9107-20, 2003 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-14647446

RESUMO

Calicheamicin thetaII is a member of the enediyne class of antitumor antibiotics that bind to DNA and induce apoptosis. These compounds differ, however, from conventional anticancer drugs as they bind in a sequence-specific manner noncovalently to DNA and cause sequence-selective oxidation of deoxyriboses and bending of the DNA helix. Calicheamicin is clinically employed as immunoconjugate to antibodies directed against, for example, CD33 in the case of gemtuzumab ozogamicin. Here, we show by the use of the unconjugated drug that calicheamicin-induced apoptosis is independent from death-receptor/FADD-mediated signals. Moreover, calicheamicin triggers apoptosis in a p53-independent manner as shown by the use of p53 knockout cells. Cell death proceeds via activation of mitochondrial permeability transition, cytochrome c release and activation of caspase-9 and -3. The overexpression of Bcl-x(L) or Bcl-2 strongly inhibited calicheamicin-induced apoptosis. Knockout of Bax abrogated cell death after calicheamicin treatment. Thus, the activation of mitochondria and execution of cell death occur through a fully Bax-dependent mechanism. Interestingly, caspase inhibition by the pancaspase-inhibitor zVAD-fmk interfered with mitochondrial activation by calicheamicin. This places caspase activation upstream of the mitochondria and indicates that calicheamicin-triggered apoptosis is enhanced through death receptor-independent activation of the caspase cascade, that is, an amplification loop that is required for full activation of the mitochondrial pathway.


Assuntos
Aminoglicosídeos/toxicidade , Antibióticos Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Mitocôndrias/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas Proto-Oncogênicas/metabolismo , Pirimidinas/toxicidade , Fragmentação do DNA , Enedi-Inos , Humanos , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/fisiologia , Mitocôndrias/efeitos dos fármacos , Permeabilidade , Fosfatidilserinas/farmacologia , Transfecção , Células Tumorais Cultivadas , Proteína X Associada a bcl-2 , Receptor fas/fisiologia
11.
ACS Chem Neurosci ; 6(3): 403-16, 2015 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-25561023

RESUMO

Parkinson's disease (PD) is associated with the formation of toxic α-synuclein oligomers that can penetrate the cell membrane. Familial forms of PD are caused by the point mutations A53T, A30P, E46K, and H50Q. Artificial point mutations E35K and E57K also increase oligomerization and pore formation. We generated structural conformations of α-synuclein and the above-mentioned mutants using molecular dynamics. We elucidated four main regions in these conformers contacting the membrane and found that the region including residues 39-45 (Zone2) may have maximum membrane penetration. E57K mutant had the highest rate of interaction with the membrane, followed by A53T, E46K, and E35K mutants and wild type (wt) α-synuclein. The mutant A30P had the smallest percentage of conformers that contact the membrane by Zone 2 than all other mutants and wt α-synuclein. These results were confirmed experimentally in vitro. We identified the key amino acids that can interact with the membrane (Y38, E62, and N65 (first hydrophilic layer); E104, E105, and D115 (second hydrophilic layer), and V15 and V26 (central hydrophobic layer)) and the residues that are involved in the interprotein contacts (L38, V48, V49, Q62, and T64). Understanding the molecular interactions of α-synuclein mutants is important for the design of compounds blocking the formation of toxic oligomers.


Assuntos
Membrana Celular/metabolismo , Mutação Puntual/genética , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Animais , Linhagem Celular Tumoral , Simulação por Computador , Humanos , Imageamento por Ressonância Magnética , Modelos Moleculares , Neuroblastoma/patologia , Dinâmica não Linear , Conformação Proteica , Estrutura Terciária de Proteína/genética , Ratos , Transfecção , alfa-Sinucleína/química
12.
Br J Pharmacol ; 171(24): 5757-73, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25117211

RESUMO

BACKGROUND AND PURPOSE: Anti-retrovirals have improved and extended the life expectancy of patients with HIV. However, as this population ages, the prevalence of cognitive changes is increasing. Aberrant activation of kinases, such as receptor tyrosine kinases (RTKs) and cyclin-dependent kinase 5 (CDK5), play a role in the mechanisms of HIV neurotoxicity. Inhibitors of CDK5, such as roscovitine, have neuroprotective effects; however, CNS penetration is low. Interestingly, tyrosine kinase inhibitors (TKIs) display some CDK inhibitory activity and ability to cross the blood-brain barrier. EXPERIMENTAL APPROACH: We screened a small group of known TKIs for a candidate with additional CDK5 inhibitory activity and tested the efficacy of the candidate in in vitro and in vivo models of HIV-gp120 neurotoxicity. KEY RESULTS: Among 12 different compounds, sunitinib inhibited CDK5 with an IC50 of 4.2 µM. In silico analysis revealed that, similarly to roscovitine, sunitinib fitted 6 of 10 features of the CDK5 pharmacophore. In a cell-based model, sunitinib reduced CDK5 phosphorylation (pCDK5), calpain-dependent p35/p25 conversion and protected neuronal cells from the toxic effects of gp120. In glial fibrillary acidic protein-gp120 transgenic (tg) mice, sunitinib reduced levels of pCDK5, p35/p25 and phosphorylated tau protein, along with amelioration of the neurodegenerative pathology. CONCLUSIONS AND IMPLICATIONS: Compounds such as sunitinib with dual kinase inhibitory activity could ameliorate the cognitive impairment associated with chronic HIV infection of the CNS. Moreover, repositioning existing low MW compounds holds promise for the treatment of patients with neurodegenerative disorders.


Assuntos
Complexo AIDS Demência , Antineoplásicos/farmacologia , Quinase 5 Dependente de Ciclina/efeitos dos fármacos , Proteína gp120 do Envelope de HIV/toxicidade , Indóis/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirróis/farmacologia , Animais , Dasatinibe , Cloridrato de Erlotinib , Flavonoides/farmacologia , Proteína gp120 do Envelope de HIV/genética , Técnicas In Vitro , Lapatinib , Camundongos , Camundongos Transgênicos , Doenças Neurodegenerativas , Purinas/farmacologia , Pirimidinas/farmacologia , Quinazolinas/farmacologia , Ratos , Roscovitina , Sunitinibe , Tiazóis/farmacologia
13.
Photochem Photobiol ; 89(3): 698-708, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23311544

RESUMO

Sparing sensitive healthy tissue from chemotherapy exposure is a critical challenge in the treatment of cancer. The work described here demonstrates the localized in vivo photoactivation of a new chemotherapy prodrug of doxorubicin (DOX). The DOX prodrug (DOX-PCB) was 200 times less toxic than DOX and was designed to release pure DOX when exposed to 365 nm light. This wavelength was chosen because it had good tissue penetration through a 1 cm diameter tumor, but had very low skin penetration, due to melanin absorption, preventing uncontrolled activation from outside sources. The light was delivered specifically to the tumor tissue using a specialized fiber-optic LED system. Pharmacokinetic studies showed that DOX-PCB had an α circulation half-life of 10 min which was comparable to that of DOX at 20 min. DOX-PCB demonstrated resistance to metabolic cleavage ensuring that exposure to 365 nm light was the main mode of in vivo activation. Tissue extractions from tumors exposed to 365 nm light in vivo showed the presence of DOX-PCB as well as activated DOX. The exposed tumors had six times more DOX concentration than nearby unexposed control tumors. This in vivo proof of concept demonstrates the first preferential activation of a photocleavable prodrug in deep tumor tissue.


Assuntos
Doxorrubicina/análogos & derivados , Doxorrubicina/farmacologia , Neoplasias Pulmonares/metabolismo , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Pró-Fármacos/farmacologia , Animais , Linhagem Celular Tumoral , Ciclodextrinas/química , Doxorrubicina/farmacocinética , Esquema de Medicação , Feminino , Humanos , Luz , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fármacos Fotossensibilizantes/síntese química , Fármacos Fotossensibilizantes/farmacocinética , Pró-Fármacos/síntese química , Pró-Fármacos/farmacocinética , Carga Tumoral/efeitos dos fármacos
14.
J Mass Spectrom ; 48(7): 768-73, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23832932

RESUMO

The localized conversion of inactive doxorubicin prodrug chemotherapeutics to pharmacalogically active forms is difficult to quantify in mouse tumor models because it occurs only in small regions of tissue. The tumor tissue extraction protocol and LC-MS/MS analysis method described here were optimized to obtain a detection limit of 7.8 pg for the activated doxorubicin and 0.36 ng for the doxorubicin prodrug. This method can be useful for determining the biodistribution and activation efficiency for many different doxorubicin prodrugs. It can also be used for quantification of doxorubicin from tumor models that have poor vascularization resulting in low tissue accumulation.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Doxorrubicina/análise , Doxorrubicina/farmacocinética , Neoplasias Pulmonares/metabolismo , Pró-Fármacos/análise , Pró-Fármacos/farmacocinética , Espectrometria de Massas em Tandem/métodos , Animais , Doxorrubicina/isolamento & purificação , Epirubicina/análise , Feminino , Limite de Detecção , Modelos Lineares , Neoplasias Pulmonares/química , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Camundongos Nus , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Int J Nanomedicine ; 8: 3991-4006, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24174874

RESUMO

Staurosporine (STS) is a potent pan-kinase inhibitor with marked activity against several chemotherapy-resistant tumor types in vitro. The translational progress of this compound has been hindered by poor pharmacokinetics and toxicity. We sought to determine whether liposomal encapsulation of STS would enhance antitumor efficacy and reduce toxicity, thereby supporting the feasibility of further preclinical development. We developed a novel reverse pH gradient liposomal loading method for STS, with an optimal buffer type and drug-to-lipid ratio. Our approach produced 70% loading efficiency with good retention, and we provide, for the first time, an assessment of the in vivo antitumor activity of STS. A low intravenous dose (0.8 mg/kg) inhibited U87 tumors in a murine flank model. Biodistribution showed preferential tumor accumulation, and body weight data, a sensitive index of STS toxicity, was unaffected by liposomal STS, but did decline with the free compound. In vitro experiments revealed that liposomal STS blocked Akt phosphorylation, induced poly(ADP-ribose) polymerase cleavage, and produced cell death via apoptosis. This study provides a basis to explore further the feasibility of liposomally encapsulated STS, and potentially related compounds for the management of resistant solid tumors.


Assuntos
Lipossomos/administração & dosagem , Lipossomos/química , Nanocápsulas/administração & dosagem , Nanocápsulas/química , Neoplasias Experimentais/tratamento farmacológico , Proteína Quinase C/antagonistas & inibidores , Estaurosporina/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Difusão , Feminino , Humanos , Lipossomos/ultraestrutura , Masculino , Camundongos , Camundongos Nus , Nanocápsulas/ultraestrutura , Neoplasias Experimentais/patologia , Estaurosporina/química , Resultado do Tratamento
16.
Bioorg Med Chem Lett ; 12(4): 557-60, 2002 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-11844671

RESUMO

Two 4'-propylcarbonoxy derivatives (2,3) of etoposide (1), a topoisomerase II inhibitor, were synthesized and evaluated as potential prodrugs for anticancer therapy. Their activation via hydrolysis mechanisms was determined as a function of pH in buffer solutions, in human serum and in the presence of carboxyl ester hydrolase. Cytotoxicity was determined on various tumor cell lines and compared to the parent compound. On cell lines exhibiting resistance to etoposide we observed an enhanced cytotoxicity of the prodrugs of up to three orders of magnitude.


Assuntos
Antineoplásicos Fitogênicos/síntese química , Etoposídeo/análogos & derivados , Pró-Fármacos/síntese química , Animais , Antineoplásicos Fitogênicos/farmacocinética , Antineoplásicos Fitogênicos/farmacologia , Carboxilesterase , Hidrolases de Éster Carboxílico/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Esterases/metabolismo , Etoposídeo/síntese química , Etoposídeo/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Hidrólise , Concentração Inibidora 50 , Inibidores da Síntese de Ácido Nucleico/síntese química , Inibidores da Síntese de Ácido Nucleico/farmacocinética , Inibidores da Síntese de Ácido Nucleico/farmacologia , Pró-Fármacos/farmacocinética , Pró-Fármacos/farmacologia , Suínos , Células Tumorais Cultivadas/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA