Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 195
Filtrar
1.
Sci Signal ; 13(631)2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32398349

RESUMO

Receptor-interacting protein kinase 1 (RIPK1) is a serine/threonine kinase that dictates whether cells survive or die in response to the cytokine tumor necrosis factor (TNF) and other inflammatory stimuli. The activity of RIPK1 is tightly controlled by multiple posttranslational modification mechanisms, including ubiquitination and phosphorylation. Here, we report that sensitivity to TNF-induced, RIPK1-dependent cell death was tunable by the pH environment. We found that an acidic extracellular pH, which led to a concomitant decrease in intracellular pH, impaired the kinase activation of RIPK1 and autophosphorylation at Ser166 Consequently, formation of the cytosolic death-inducing complex II and subsequent RIPK1-dependent necroptosis and apoptosis were inhibited. By contrast, low pH did not affect the formation of membrane-anchored TNFR1-containing signaling complex (complex I), RIPK1 ubiquitination, and NF-κB activation. TNF-induced cell death in Ripk1 -/- cells was not sensitive to pH changes. Furthermore, mutation of the conserved His151 abolished the pH dependence of RIPK1 activation, suggesting that this histidine residue functions as a proton acceptor to modulate RIPK1 activity in response to pH changes. These results revealed an unexpected environmental factor that controls the death-inducing activity of RIPK1.


Assuntos
Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Células 3T3 , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Fator de Necrose Tumoral alfa/farmacocinética
2.
Mol Cell Biochem ; 464(1-2): 83-91, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31741130

RESUMO

Kawasaki disease is an immune-mediated acute, systemic vasculitis and is the leading cause of acquired heart disease in children in the developed world. Bifidobacterium (BIF) is one of the dominant bacteria in the intestines of humans and many mammals and is able to adjust the intestinal flora disorder. The Caco-2 cell monolayers were treated with tumor necrosis factor-α (TNF-α) at 10 ng/ml for 24 h to induce the destruction of intestinal mucosal barrier system. Cells viability was detected through Cell Counting Kit-8 assay. Cell apoptosis was measured by flow cytometry and the expression of apoptosis related proteins was also detected through Western blot. The level of pro-inflammatory cytokines interleukin-6 (IL-6) and IL-8 was detected through ELISA, Western blot and qRT-PCR, respectively. Transepithelial electrical resistance (TEER) assay was conducted to value the barrier function of intestinal mucosa. Cell autophagy and NF-κB and p38MAPK pathways associated proteins were examined through Western blot. In the absence of TNF-α treatment, cell viability and apoptosis showed no significant change. TNF-α decreased cell viability and increased cell apoptosis and BIF treatment mitigated the TNF-α-induced change. Then, we found that BIF treatment effectively suppressed TNF-α-induced overexpression of IL-6 and IL-8. Besides, the results of TEER assay showed that barrier function of intestinal mucosa which was destroyed by TNF-α was effectively recovered by BIF treatment. In addition, TNF-α induced autophagy was also suppressed by BIF. Moreover, TNF-α activated NF-κB and p38MAPK signal pathways were also blocked by BIF, SN50 and SB203580. Our present study reveals that BIF plays a protective role in TNF-α-induced inflammatory response in Caco-2 cells through NF-κB and p38MAPK pathways.


Assuntos
Bifidobacterium , Mucosa Intestinal/metabolismo , Sistema de Sinalização das MAP Quinases , Síndrome de Linfonodos Mucocutâneos/prevenção & controle , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Células CACO-2 , Humanos , Inflamação/metabolismo , Inflamação/patologia , Inflamação/prevenção & controle , Mucosa Intestinal/patologia , Síndrome de Linfonodos Mucocutâneos/metabolismo , Síndrome de Linfonodos Mucocutâneos/patologia , Fator de Necrose Tumoral alfa/farmacocinética
3.
Blood ; 134(3): 252-262, 2019 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-31118164

RESUMO

Patients with primary central nervous system lymphoma (PCNSL) are treated with high-dose methotrexate-based chemotherapy, which requires hospitalization and extensive expertise to manage related toxicity. The use of R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone) could overcome these difficulties, but blood-brain barrier (BBB) penetration of related drugs is poor. Tumor necrosis factor-α coupled with NGR (NGR-hTNF), a peptide targeting CD13+ vessels, induces endothelial permeabilization and improves tumor access of cytostatics. We tested the hypothesis that NGR-hTNF can break the BBB, thereby improving penetration and activity of R-CHOP in patients with relapsed/refractory PCNSL (NCT03536039). Patients received six R-CHOP21 courses, alone at the first course and preceded by NGR-hTNF (0.8 µg/m2) afterward. This trial included 2 phases: an "explorative phase" addressing the effect of NGR-hTNF on drug pharmacokinetic parameters and on vessel permeability, assessed by dynamic contrast-enhanced magnetic resonance imaging and 99mTc-diethylene-triamine-pentacetic acid-single-photon emission computed tomography, and the expression of CD13 on tumor tissue; and an "expansion phase" with overall response rate as the primary end point, in which the 2-stage Simon Minimax design was used. At the first stage, if ≥4 responses were observed among 12 patients, the study accrual would have continued (sample size, 28). Herein, we report results of the explorative phase and the first-stage analysis (n = 12). CD13 was expressed in tumor vessels of all cases. NGR-hTNF selectively increased vascular permeability in tumoral/peritumoral areas, without interfering with drug plasma/cerebrospinal fluid concentrations. The NGR-hTNF/R-CHOP combination was well tolerated: there were only 2 serious adverse events, and grade 4 toxicity was almost exclusively hematological, which were resolved without dose reductions or interruptions. NGR-hTNF/R-CHOP was active, with 9 confirmed responses (75%; 95% confidence interval, 51-99), 8 of which were complete. In conclusion, NGR-hTNF/R-CHOP was safe in these heavily pretreated patients. NGR-hTNF enhanced vascular permeability specifically in tumoral/peritumoral areas, which resulted in fast and sustained responses.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Barreira Hematoencefálica/efeitos dos fármacos , Neoplasias do Sistema Nervoso Central/tratamento farmacológico , Linfoma não Hodgkin/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacocinética , Fator de Necrose Tumoral alfa/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Biomarcadores , Barreira Hematoencefálica/diagnóstico por imagem , Antígenos CD13/metabolismo , Permeabilidade da Membrana Celular , Neoplasias do Sistema Nervoso Central/diagnóstico , Neoplasias do Sistema Nervoso Central/metabolismo , Neoplasias do Sistema Nervoso Central/mortalidade , Ciclofosfamida/efeitos adversos , Ciclofosfamida/uso terapêutico , Doxorrubicina/efeitos adversos , Doxorrubicina/uso terapêutico , Feminino , Humanos , Imuno-Histoquímica , Linfoma não Hodgkin/diagnóstico , Linfoma não Hodgkin/metabolismo , Linfoma não Hodgkin/mortalidade , Masculino , Neuroimagem/métodos , Prednisona/efeitos adversos , Prednisona/uso terapêutico , Proteínas Recombinantes de Fusão/administração & dosagem , Projetos de Pesquisa , Rituximab/efeitos adversos , Rituximab/uso terapêutico , Tomografia Computadorizada de Emissão de Fóton Único , Resultado do Tratamento , Fator de Necrose Tumoral alfa/administração & dosagem , Vincristina/efeitos adversos , Vincristina/uso terapêutico
4.
J Biol Chem ; 293(52): 20062-20072, 2018 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-30361438

RESUMO

The linear ubiquitin chain assembly complex (LUBAC) regulates NF-κB activation by modifying proteins with linear (M1-linked) ubiquitination chains. Although LUBAC also regulates the apoptosis pathway, the precise mechanism by which LUBAC regulates apoptosis remains not fully defined. Here, we report that LUBAC-mediated M1-linked ubiquitination of cellular FLICE-like inhibitory protein (cFLIP), an anti-apoptotic molecule, contributes to tumor necrosis factor (TNF) α-induced apoptosis. We found that deficiency of RNF31, the catalytic subunit of the LUBAC complex, promoted cFLIP degradation in a proteasome-dependent manner. Moreover, we observed RNF31 directly interact with cFLIP, and LUBAC further conjugated M1-linked ubiquitination chains at Lys-351 and Lys-353 of cFLIP to stabilize cFLIP, thereby protecting cells from TNFα-induced apoptosis. Together, our study identifies a new substrate of LUBAC and reveals a new molecular mechanism through which LUBAC regulates TNFα-induced apoptosis via M1-linked ubiquitination.


Assuntos
Apoptose/efeitos dos fármacos , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Complexos Multienzimáticos/metabolismo , Fator de Necrose Tumoral alfa/farmacocinética , Ubiquitinação/efeitos dos fármacos , Apoptose/genética , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Células HEK293 , Células HeLa , Humanos , Complexos Multienzimáticos/genética , Ubiquitinação/genética
5.
Adv Healthc Mater ; 6(13)2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28557249

RESUMO

Tumor necrosis factor-alpha (TNF-α) is a pleiotropic immune stimulatory cytokine and natural endotoxin that can induce necrosis and regression in solid tumors. However, systemic administration of TNF-α is not feasible due to its short half-life and acute toxicity, preventing its widespread use in cancer treatment. Dendritic mesoporous silica nanoparticles (DMSN) are used coated with a pH-responsive block copolymer gate system combining charged hyperbranched polyethylenimine and nonionic hydrophilic polyethylenglycol to encapsulate TNF-α and deliver it into various cancer cell lines and dendritic cells. Half-maximal effective concentration (EC50 ) for loaded TNF-α is reduced by more than two orders of magnitude. Particle stability and premature cargo release are assessed with enzyme-linked immunosorbent assay. TNF-α-loaded particles are stable for up to 5 d in medium. Tumor cells are grown in vitro as 3D fluorescent ubiquitination-based cell cycle indicator spheroids that mimic in vivo tumor architecture and microenvironment, allowing real-time cell cycle imaging. DMSN penetrate these spheroids, release TNF-α from its pores, preferentially affect cells in S/G2/M phase, and induce cell death in a time- and dose-dependent manner. In conclusion, DMSN encapsulation is demonstrated, which is a promising approach to enhance delivery and efficacy of antitumor drugs, while minimizing adverse side effects.


Assuntos
Ciclo Celular/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas , Neoplasias/tratamento farmacológico , Dióxido de Silício , Fator de Necrose Tumoral alfa , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Nanopartículas/química , Nanopartículas/uso terapêutico , Neoplasias/metabolismo , Porosidade , Dióxido de Silício/química , Dióxido de Silício/farmacocinética , Dióxido de Silício/farmacologia , Fator de Necrose Tumoral alfa/química , Fator de Necrose Tumoral alfa/farmacocinética , Fator de Necrose Tumoral alfa/farmacologia
6.
Drug Metab Dispos ; 45(7): 798-806, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28411279

RESUMO

Tumor necrosis factor-α (TNF-α) is a soluble cytokine and target of specific monoclonal antibodies (mAbs) and other biologic agents used in the treatment of inflammatory diseases. These biologics exert their pharmacological effects through binding and neutralizing TNF-α, and thus they prevent TNF-α from interacting with its cell surface receptors. The magnitude of the pharmacological effects is governed not only by the pharmacokinetics (PK) of mAbs, but also by the kinetic fate of TNF-α We have examined the pharmacokinetics of recombinant human TNF-α (rhTNF-α) in rats at low doses and quantitatively characterized its pharmacokinetic features with a minimal physiologically based pharmacokinetic model. Our experimental and literature-digitalized PK data of rhTNF-α in rats across a wide range of doses were applied to global model fitting. rhTNF-α exhibits permeability rate-limited tissue distribution and its elimination is comprised of a saturable clearance pathway mediated by tumor necrosis factor receptor binding and disposition and renal filtration. The resulting model integrated with classic allometry was further used for interspecies PK scaling and resulted in model predictions that agreed well with experimental measurements in monkeys. In addition, a semimechanistic model was proposed and applied to explore the absorption kinetics of rhTNF-α following s.c. and other routes of administration. The model suggests substantial presystemic degradation of rhTNF-α for s.c. and i.m. routes and considerable lymph uptake contributing to the overall systemic absorption through the stomach wall and gastrointestinal wall routes of dosing. This report provides comprehensive modeling and key insights into the complexities of absorption and disposition of a major cytokine.


Assuntos
Modelos Biológicos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacocinética , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/farmacocinética , Absorção Fisiológica , Animais , Relação Dose-Resposta a Droga , Haplorrinos , Infusões Subcutâneas , Injeções Intravenosas , Injeções Subcutâneas , Masculino , Especificidade de Órgãos , Ratos Endogâmicos Lew , Proteínas Recombinantes/sangue , Especificidade da Espécie , Distribuição Tecidual , Fator de Necrose Tumoral alfa/sangue
7.
Aliment Pharmacol Ther ; 45(10): 1329-1338, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28318043

RESUMO

BACKGROUND: Transplacental transfer of infliximab and adalimumab results in detectable drug levels in the cord blood and infant. AIM: To determine if pregnancy influenced the pharmacokinetics of anti-TNF agents in women with inflammatory bowel disease. METHODS: Twenty-five women from the University of Calgary inflammatory bowel disease(IBD) pregnancy clinic on maintenance infliximab or adalimumab were recruited prospectively with serum bio-banking performed each trimester. Infliximab trough and adalimumab steady-state levels were the outcomes of interest and were analysed using the ANSER infliximab and adalimumab assays. Multivariate linear mixed-effects models were constructed to assess infliximab and adalimumab drug levels during pregnancy adjusting for the clinical covariates of albumin, BMI and CRP. RESULTS: Fifteen women (eight Crohn's disease, seven ulcerative colitis) received infliximab and 10 women with 11 pregnancies were treated with adalimumab. Median age was 29.6 years (IQR: 27.6-31.2 years). Median disease duration was 9.2 years (IQR: 3.16-15.0 years). Median trough infliximab concentrations were 8.50 µg/mL (IQR: 7.23-10.07 µg/mL), 10.31 µg/mL (IQR: 7.66-15.63 µg/mL) and 21.02 µg/mL (IQR: 16.01-26.70 µg/mL) at trimesters 1, 2 and 3 respectively. Significant changes in albumin and BMI (P < 0.05) but not CRP (P > 0.05) were documented throughout pregnancy. After adjusting for albumin, BMI and CRP, infliximab trough levels increased during pregnancy, by 4.2 µg/mL per trimester (P = 0.02), while adalimumab drug levels remained stable (P > 0.05). CONCLUSIONS: Infliximab levels rise during pregnancy, whereas adalimumab levels remain stable after accounting for changes in albumin, BMI and CRP. Therapeutic drug monitoring in the second trimester may be useful in guiding dosing in the third trimester.


Assuntos
Adalimumab/metabolismo , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/metabolismo , Infliximab/farmacocinética , Troca Materno-Fetal , Adalimumab/farmacologia , Adalimumab/uso terapêutico , Adolescente , Adulto , Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais Humanizados/uso terapêutico , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/metabolismo , Doença de Crohn/tratamento farmacológico , Doença de Crohn/metabolismo , Monitoramento de Medicamentos , Feminino , Humanos , Infliximab/uso terapêutico , Troca Materno-Fetal/efeitos dos fármacos , Placenta/efeitos dos fármacos , Placenta/metabolismo , Circulação Placentária , Gravidez , Fator de Necrose Tumoral alfa/farmacocinética , Fator de Necrose Tumoral alfa/uso terapêutico , Adulto Jovem
8.
Adv Mater ; 29(13)2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28160337
9.
Biochemistry (Mosc) ; 81(10): 1188-1197, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27908243

RESUMO

Prolonged or excessive increase in the circulatory level of proinflammatory tumor necrosis factor (TNF) leads to abnormal activation and subsequent damage to endothelium. TNF at high concentrations causes apoptosis of endothelial cells. Previously, using mitochondria-targeted antioxidants of SkQ family, we have shown that apoptosis of endothelial cells is dependent on the production of reactive oxygen species (ROS) in mitochondria (mito-ROS). Now we have found that TNF at low concentrations does not cause cell death but activates caspase-3 and caspase-dependent increase in endothelial permeability in vitro. This effect is probably due to the cleavage of ß-catenin - an adherent junction protein localized in the cytoplasm. We have also shown that extracellular matrix metalloprotease 9 (MMP9) VE-cadherin shedding plays a major role in the TNF-induced endothelial permeability. The mechanisms of the caspase-3 and MMP9 activation are probably not related to each other since caspase inhibition did not affect VE-cadherin cleavage and MMP9 inhibition had no effect on the caspase-3 activation. Mitochondria-targeted antioxidant SkQR1 inhibited TNF-induced increase in endothelial permeability. SkQR1 also inhibited caspase-3 activation, ß-catenin cleavage, and MMP9-dependent VE-cadherin shedding. The data suggest that mito-ROS are involved in the increase in endothelial permeability due to the activation of both caspase-dependent cleavage of intracellular proteins and of MMP9-dependent cleavage of the transmembrane cell-to-cell contact proteins.


Assuntos
Antioxidantes/farmacologia , Permeabilidade Capilar/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Plastoquinona/análogos & derivados , Rodaminas/farmacologia , Fator de Necrose Tumoral alfa/farmacocinética , Antígenos CD/metabolismo , Apoptose/efeitos dos fármacos , Caderinas/metabolismo , Caspase 3/metabolismo , Caspase 9/metabolismo , Linhagem Celular , Células Endoteliais/citologia , Endotélio Vascular/citologia , Humanos , Mitocôndrias/metabolismo , Plastoquinona/farmacologia
10.
Drug Deliv ; 23(6): 2044-57, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26599817

RESUMO

Enteric coating of microparticles prevents stomach degradation and enhances oral bioavailability of poorly soluble drugs. Thus, in the present study, enteric-coated microparticle (ECMP) of biochanin A was developed and their pharmacological potential was investigated in hypertensive ovariectomized rats (Ovx-HT). Biochanin A microparticles were prepared by using ionic gelation method and coating was done by ethyl cellulose using coacervation phase separation method. Surface modified microparticles were characterized on the basis of size, entrapment efficiency, polydispersity index, FTIR and zeta potential and percentage of in vitro release. Ovariectomized rats were administered deoxy corticosterone acetate (40 mg/kg, s.c. twice a week for 6 weeks) salt to induce Ovx-HT. Hypertension was assessed in terms of increase in systolic, diastolic, mean arterial blood pressure, lipid peroxidation level, tumor necrosis factor-alpha (TNF-α) level and decrease in serum nitrite and reduced glutathione (GSH) level. The optimized formulation of ECMP has shown significant increase in oral bioavailability assessed by high-performance liquid chromatography. Furthermore, these ECMPs significantly reduced the mean arterial blood pressure, systolic and diastolic blood pressure, reduced lipid peroxidation and TNF-α level and significantly increased the serum nitrite and reduced GSH level in Ovx-HT rats. However, L-NAME significantly prevented the ameliorative effect of ECMP. Thus, it may be concluded that ECMP of biochanin A has shown delayed release capacity, increase in oral bioavailability up to 6 folds than plain biochanin A and exerts antihypertensive effect in ovariectomized rats possibly in an eNOS dependent manner.


Assuntos
Anti-Hipertensivos/química , Portadores de Fármacos/química , Estrogênios/química , Genisteína/química , Genisteína/farmacologia , Hipertensão/tratamento farmacológico , Fator de Necrose Tumoral alfa/química , Animais , Anti-Hipertensivos/farmacocinética , Disponibilidade Biológica , Química Farmacêutica , Genisteína/farmacocinética , Hipertensão/metabolismo , Ratos , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacocinética
11.
Sci Rep ; 5: 18363, 2015 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-26678960

RESUMO

Chemical conjugation is commonly used to enhance the pharmacokinetics, biodistribution, and potency of protein therapeutics, but often leads to non-specific modification or loss of bioactivity. Here, we present a simple, versatile and widely applicable method that allows exquisite N-terminal specific modification of proteins. Combining reversible side-chain blocking and protease mediated cleavage of a commonly used HIS tag appended to a protein, we generate with high yield and purity exquisitely site specific and selective bio-conjugates of TNF-α by using amine reactive NHS ester chemistry. We confirm the N terminal selectivity and specificity using mass spectral analyses and show near complete retention of the biological activity of our model protein both in vitro and in vivo murine models. We believe that this methodology would be applicable to a variety of potentially therapeutic proteins and the specificity afforded by this technique would allow for rapid generation of novel biologics.


Assuntos
Química Farmacêutica/métodos , Fator de Necrose Tumoral alfa/química , Aminas/química , Animais , Linhagem Celular , Feminino , Meia-Vida , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Polietilenoglicóis/química , Estrutura Terciária de Proteína , Succinimidas/química , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/farmacocinética
12.
J Rheumatol ; 42(12): 2376-82, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26568593

RESUMO

OBJECTIVE: The purpose of this study was to (1) evaluate baseline characteristics of nonradiographic axial spondyloarthritis (nr-axSpA) and ankylosing spondylitis (AS) treated with tumor necrosis factor-α inhibitors (TNFi), (2) assess the response to first TNFi treatment, and (3) compare drug-survival duration and rates. METHODS: Inclusion criteria were patients with axSpA who initiated first TNFi treatment between April 2001 and July 2014 and were followed up for at least 3 months. Efficacy criteria were an improvement of at least 2 points (on a 0-10 scale) or a 50% improvement in the Bath Ankylosing Spondylitis Disease Activity Index (BASDAI). Baseline characteristics, responses at 12 months, and drug survival were compared between AS and nr-axSpA. RESULTS: A total of 361 patients were included in the study (AS, n = 263 and nr-axSpA, n = 98). Patients with AS were more often men (65.02% vs 45.92%, p = 0.001) and had longer symptom duration (11.71 ± 9.52 vs 7.34 ± 9.30 yrs, p < 0.001). Median levels of acute-phase reactants (C-reactive protein and erythrocyte sedimentation rate) were significantly higher in patients with AS (p < 0.001 for both). Median BASDAI scores at first TNFi initiation were not higher in patients with nr-axSpA than in patients with AS (59, 49-70 vs 60, 50-70, p = 0.73). BASDAI 20 and BASDAI 50 response rates at 12 months were not statistically different between patients with AS and patients with nr-axSpA (74.58% vs 64.58%, p = 0.19 and 61.02% vs 50.00%, p = 0.19, respectively). No statistically significant difference in terms of survival was observed between patients with AS and nr-axSpA (p = 1.00). CONCLUSION: Treatment response and drug survival were similar in patients with AS and nr-axSpA after first TNFi initiation.


Assuntos
Espondilartrite/diagnóstico , Espondilartrite/tratamento farmacológico , Espondilite Anquilosante/diagnóstico , Espondilite Anquilosante/tratamento farmacológico , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Adulto , Fatores Etários , Estudos de Coortes , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Seguimentos , Humanos , Estimativa de Kaplan-Meier , Estudos Longitudinais , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Medição da Dor , Estudos Retrospectivos , Medição de Risco , Índice de Gravidade de Doença , Fatores Sexuais , Resultado do Tratamento , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/farmacocinética
13.
Gastroenterology ; 149(2): 350-5.e2, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25917786

RESUMO

BACKGROUND & AIMS: It is not clear why some patients with ulcerative colitis (UC) do not respond to treatment with anti-tumor necrosis factor (TNF) agents, such as infliximab. It could be that some patients have high level of inflammation, with large quantities of TNF to be neutralized by the drug. We investigated whether loss of anti-TNF agents through ulcerated intestinal mucosa reduces the efficacy of these drugs in patients with severe UC. METHODS: We collected fecal samples from 30 consecutive patients with moderate to severely active UC during the first 2 weeks of infliximab therapy at the University of Amsterdam hospital. Infliximab concentrations were measured in serum and supernatants of fecal samples using an enzyme-linked immunosorbent assay (Sanquin Biologicals Laboratory, Amsterdam, The Netherlands). Clinical and endoscopic responses were assessed 2 and 8 weeks and 3 months after treatment began. RESULTS: Infliximab was detected in 129 of 195 fecal samples (66%); the highest concentrations were measured in the first days after the first infusion. Patients that were clinical nonresponders at week 2 had significantly higher fecal concentrations of infliximab after the first day of treatment than patients with clinical responses (median concentration, 5.01 µg/mL in nonresponders vs 0.54 µg/mL in responders; P = .0047). We did not observe a correlation between fecal and serum concentrations of infliximab. CONCLUSIONS: Infliximab is lost into stools of patients with UC. High fecal concentrations of infliximab in the first days after therapy begins are associated with primary nonresponse. Additional studies are needed to determine how therapeutic antibodies are lost through the intestinal mucosa and how this process affects treatment response. Clinical trial ID: NL41310.018.12.


Assuntos
Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/uso terapêutico , Colite Ulcerativa/tratamento farmacológico , Fezes/química , Mucosa Intestinal/efeitos dos fármacos , Adulto , Anticorpos Monoclonais/administração & dosagem , Colite Ulcerativa/sangue , Colite Ulcerativa/patologia , Monitoramento de Medicamentos/métodos , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Infliximab , Mucosa Intestinal/patologia , Masculino , Pessoa de Meia-Idade , Países Baixos , Índice de Gravidade de Doença , Resultado do Tratamento , Fator de Necrose Tumoral alfa/farmacocinética , Fator de Necrose Tumoral alfa/uso terapêutico
14.
Acta Biomater ; 12: 174-182, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25305513

RESUMO

Liver fibrosis is a chronic liver disease caused by viral infection and/or metabolic, genetic and cholestatic disorders. The inhibition of hepatic stellate cell (HSC) activation and the selective apoptosis of activated HSCs can be a good strategy to treat liver fibrosis. The activated HSCs are known to be more susceptible to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induced apoptosis than normal HSCs because death receptor 5 is overexpressed on the cell surface. In this work, a target-specific and long-acting hyaluronic acid (HA)-TRAIL conjugate was successfully developed for the treatment of liver fibrosis. The HA-TRAIL conjugate was synthesized by a coupling reaction between aldehyde-modified HA and the N-terminal amine group of TRAIL. The biological activity of the HA-TRAIL conjugate was confirmed by an in vitro anti-proliferation assay and caspase-3 expression in human colon cancer HCT116 cells. In vivo real-time bioimaging exhibited the target-specific delivery of near-infrared fluorescence dye-labeled HA-TRAIL conjugate to the liver in mice. According to pharmacokinetic analysis, the HA-TRAIL conjugate was detected for more than 4days after single intravenous injection into Sprague-Dawley (SD) rats. Finally, we could confirm the antifibrotic effect of HA-TRAIL conjugate in an N-nitrosodimethylamine-induced liver fibrosis model SD rats.


Assuntos
Ácido Hialurônico/uso terapêutico , Cirrose Hepática/tratamento farmacológico , Fator de Necrose Tumoral alfa/uso terapêutico , Animais , Ácido Hialurônico/química , Ácido Hialurônico/farmacocinética , Ligantes , Masculino , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/química , Fator de Necrose Tumoral alfa/farmacocinética
15.
Drug Dev Ind Pharm ; 41(3): 369-74, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24274583

RESUMO

Local drug delivery strategies based on nanoparticles, gels, polymeric films, rods and wafers are increasingly used in cancer chemotherapy in order to enhance therapeutic effect and reduce systemic toxicity. Herein, a biodegradable and biocompatible in situ thermosensitive hydrogel was designed and employed to deliver tumor necrosis factor-α (TNF-α) locally by intratumoral injection. The triblock copolymer was synthesized by ring-opening polymerization (ROP) of ß-butyrolactone (ß-BL) and lactide (LA) in bulk using polyethylene glycol (PEG) as an initiator and Sn(Oct)2 as the catalyst, the polymer was characterized by NMR, gel permeation chromatography and differential scanning calorimetry. Blood and tumor pharmacokinetics and in vivo antitumor activity of TNF-α after intratumoral administration in hydrogel or solution with the same dose were evaluated on S180 tumor-bearing mice. Compared with TNF-α solution, TNF-α hydrogel exhibited a longer T1/2 (4-fold) and higher AUCtumor (19-fold), but Cmax was lower (0.5-fold), which means that the hydrogel formulation improved the efficacy with a lower systhemic exposure than the solution formation. In addition, TNF-α hydrogel improved the antitumor activity and survival due to lower systemic exposure than the solution. These results demonstrate that the in situ thermosensitive hydrogel-based local delivery system by intratumoral injection is well suited for the administration of TNF-α.


Assuntos
Antineoplásicos/administração & dosagem , Temperatura Alta/uso terapêutico , Hidrogel de Polietilenoglicol-Dimetacrilato/administração & dosagem , Injeções Intralesionais/métodos , Fator de Necrose Tumoral alfa/administração & dosagem , Animais , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacocinética , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fator de Necrose Tumoral alfa/farmacocinética
16.
J Parkinsons Dis ; 4(3): 349-60, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25061061

RESUMO

BACKGROUND: Parkinson's disease (PD) is a complex multi-system age-related neurodegenerative disorder. Targeting the ongoing neuroinflammation in PD patients is one strategy postulated to slow down or halt disease progression. Proof-of-concept studies from our group demonstrated that selective inhibition of soluble Tumor Necrosis Factor (solTNF) by intranigral delivery of dominant negative TNF (DN-TNF) inhibitors reduced neuroinflammation and nigral dopamine (DA) neuron loss in endotoxin and neurotoxin rat models of nigral degeneration. OBJECTIVE: As a next step toward human clinical trials, we aimed to determine the extent to which peripherally administered DN-TNF inhibitor XPro®1595 could: i) cross the blood-brain-barrier in therapeutically relevant concentrations, ii) attenuate neuroinflammation (microglia and astrocyte), and iii) mitigate loss of nigral DA neurons in rats receiving a unilateral 6-hydroxydopamine (6-OHDA) striatal lesion. METHODS: Rats received unilateral 6-OHDA (20 µg into the right striatum). Three or 14 days after lesion, rats were dosed with XPro®1595 (10 mg/kg in saline, subcutaneous) every third day for 35 days. Forelimb asymmetry was used to assess motor deficits after the lesion; brains were harvested 35 days after the lesion for analysis of XPro®1595 levels, glial activation and nigral DA neuron number. RESULTS: Peripheral subcutaneous dosing of XPro®1595 achieved plasma levels of 1-8 microgram/mL and CSF levels of 1-6 ng/mL depending on the time the rats were killed after final XPro®1595 injection. Irrespective of start date, XPro®1595 significantly reduced microglia and astrocyte number in SNpc whereas loss of nigral DA neurons was attenuated when drug was started 3, but not 14 days after the 6-OHDA lesion. CONCLUSIONS: Our data suggest that systemically administered XPro®1595 may have disease-modifying potential in PD patients where inflammation is part of their pathology.


Assuntos
Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/patologia , Substância Negra/efeitos dos fármacos , Substância Negra/patologia , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Barreira Hematoencefálica , Contagem de Células , Modelos Animais de Doenças , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Encefalite/prevenção & controle , Gliose/prevenção & controle , Masculino , Oxidopamina , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacocinética
17.
J Biotechnol ; 186: 1-12, 2014 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-24992210

RESUMO

Specific delivery of TNF-α antagonist to the inflamed site can increase its efficacy and reduce the side effects. In this study, we constructed a bispecific diabody (BsDb) that targets TNF-α and ED-B-containing fibronectin (B-FN), a fibronectin isoform specifically expressed in the pannus of the inflamed joint in rheumatoid arthritis. BsDb was produced in Escherichia coli as inclusion bodies, purified to homogeneity, and refolded to the functional form. Our data demonstrate that BsDb could simultaneously bind to human TNF-α and B-FN and neutralize TNF-α action. In the collagen-induced arthritis mouse model, we compared the biodistrubtion and therapeutic efficacy of BsDb with those of the anti-TNF-α scFv (TNF-scFv). Similar to TNF-scFv, BsDb penetrated into the synovial tissue quickly, showing a rapid clearance from blood and normal organs. In contrast, BsDb could selectively accumulate and retain in arthritic joints of mice for a long period time, resulting in a much stronger inhibition of arthritis progression in mice than TNF-scFv. The findings described herein indicate that BsDb has a good specificity to the inflamed joint, with low toxicity to normal organs and seems to be an ideal biological agent for the treatment of RA and other chronic inflammatory disease.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Artrite Experimental/tratamento farmacológico , Fibronectinas/uso terapêutico , Engenharia de Proteínas/métodos , Fator de Necrose Tumoral alfa/uso terapêutico , Sequência de Aminoácidos , Animais , Anticorpos Biespecíficos/química , Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/farmacocinética , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Sequência de Bases , Linhagem Celular Tumoral , Citocinas/análise , Citocinas/metabolismo , Progressão da Doença , Fibronectinas/química , Fibronectinas/genética , Fibronectinas/farmacocinética , Pé/patologia , Camundongos , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico , Distribuição Tecidual , Fator de Necrose Tumoral alfa/química , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacocinética
18.
Eur J Pharmacol ; 740: 722-32, 2014 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-24929054

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) exhibits potent antitumor activity in a wide range of cancers without deleterious side effects on normal tissues. Several TRAIL derivatives have been developed to improve its pharmacokinetics and therapeutic effects through strategies such as adding a leucine zipper to increase the circulation half-life. To obtain clinical grade LZ-TRAIL for phase I clinical trial, a single batch of 30 L bioreactor culture was performed using the Escherichia coli BL21 (DE3) strain expressing the recombinant LZ-TRAIL. A robust LZ-TRAIL production fermentation process was developed, which could be scaled up from 5L to 50 L, and had a titer of approximately 1.4 g/l. A four-step purification strategy was carried out to obtain a final product with over 95% purity and 45% yield. The final material was filter sterilized, aseptically vialed, and stored at 4°C, and comprehensively characterized using multiple assays (vialed product was sterile, purity was 95%, aggregates were <5%, potency revealed IC50 of 9 nM on MDA-MB-231 cells, and the endotoxin level was <0.25 U/mg). The purity, composition, and functional activities of the molecule were confirmed. in vivo investigations indicated that LZ-TRAIL has better antitumor potency in three Xenograft tumor models compared to TRAIL (95-281). LZ-TRAIL also showed improved pharmacokinetic and safety profiles in cynomolgus monkeys without abnormalities associated with drug exposure. In conclusion, the scalable synthesis of LZ-TRAIL is useful for production of phase I clinical trial material. These preclinical investigations warrant further clinical development of this product for cancer therapy.


Assuntos
Antineoplásicos , Glicoproteínas de Membrana , Proteínas Recombinantes de Fusão , Fator de Necrose Tumoral alfa , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ensaios Clínicos Fase I como Assunto , Escherichia coli/genética , Escherichia coli/metabolismo , Feminino , Hepatócitos/efeitos dos fármacos , Humanos , Macaca fascicularis , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/farmacocinética , Glicoproteínas de Membrana/farmacologia , Glicoproteínas de Membrana/uso terapêutico , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Ligante Indutor de Apoptose Relacionado a TNF/farmacocinética , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Carga Tumoral/efeitos dos fármacos , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/farmacocinética , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Int J Pharm ; 461(1-2): 181-91, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24269209

RESUMO

GX1 is a tumor targeting peptide. In this study, we evaluated the antitumor efficacy of a GX1-derived fusion toxin, GX1-rmhTNFα, and investigated its targeting efficiency and pharmacokinetics in vivo using multimodality imaging. Flow cytometry revealed a greater level of cell apoptosis induced by GX1-rmhTNFα (27.1%) compared with rmhTNFα or a saline control (13.7% and 4.7%, respectively). SPECT (single-photon emission computed tomography) demonstrated high accumulation of GX1-rmhTNFα in tumor site. Biodistribution studies indicated GX1-rmhTNFα was cleared by the liver and kidney, and the drug may not cross the blood-brain barrier. In addition, bioluminescence imaging (BLI) showed that GX1-rmhTNFα caused a satisfactory delay in tumor growth in both subcutaneous and orthotopic cancer models. Contrast-enhanced ultrasound (CEUS) and CD31 staining revealed a loss in blood perfusion and vasculature. TUNEL and Ki67 staining validated the in vivo results. Biochemical analyses revealed limited renal and hepatic toxicity of GX1-rmhTNFα. This study demonstrated that GX1-rmhTNFα is a safe and potent anticancer agent that may have great potential for the targeted therapy of gastric cancer.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Peptídeos Cíclicos/química , Fator de Necrose Tumoral alfa/administração & dosagem , Animais , Antineoplásicos/farmacocinética , Apoptose/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Feminino , Citometria de Fluxo , Células Endoteliais da Veia Umbilical Humana , Humanos , Marcação In Situ das Extremidades Cortadas , Antígeno Ki-67/análise , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Imagem Multimodal/métodos , Proteínas Recombinantes de Fusão , Distribuição Tecidual , Tomografia Computadorizada de Emissão de Fóton Único , Fator de Necrose Tumoral alfa/farmacocinética
20.
Sci China Life Sci ; 56(1): 51-8, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23314867

RESUMO

To design a releasable PEGylated TNF-α (rPEG-TNF-α), a cathepsin B-sensitive dipeptide (Val-Cit moiety) was inserted into conventional PEG-modified TNF-α (PEG-TNF-α), facilitating its clinical use for anti-tumor therapy. Comparative pharmacokinetic and pharmacodynamic studies showed that the half-lives of both PEGylated forms of TNF-α were ∼60-fold greater than that of unmodified TNF-α. In addition, the in vitro bioactivity of rPEG-TNF-α was greater than that of PEG-TNF-α with the same degree of PEG modification. Release of TNF-α from rPEG-TNF-α in vitro was dependent on the presence of cathepsin B and was inhibited by a cathepsin B inhibitor. Despite the potent cytotoxicity of unmodified TNF-α against normal cells, its PEGylated forms at higher TNF-α concentrations showed low cytotoxic activity against these cells. In contrast, both forms of PEGylated TNF-α showed potent cytotoxic activity against the B16 and L929 cell lines, with rPEG-TNF-α being 5- and 9-fold more potent, respectively, than PEG-TNF-α. Moreover, rPEG-TNF-α was a more potent in vivo antitumor agent than PEG-TNF-α.


Assuntos
Dipeptídeos/química , Neoplasias Experimentais/tratamento farmacológico , Polietilenoglicóis/química , Fator de Necrose Tumoral alfa/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Catepsina B/antagonistas & inibidores , Catepsina B/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Composição de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Glicopeptídeos/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Leucina/análogos & derivados , Leucina/farmacologia , Lisossomos/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Pepstatinas/farmacologia , Inibidores de Proteases/farmacologia , Ratos , Fatores de Tempo , Carga Tumoral/efeitos dos fármacos , Fator de Necrose Tumoral alfa/química , Fator de Necrose Tumoral alfa/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA