Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 208
Filter
1.
Sci Signal ; 13(631)2020 05 12.
Article in English | MEDLINE | ID: mdl-32398349

ABSTRACT

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.


Subject(s)
Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , 3T3 Cells , Animals , Cell Death/drug effects , Cell Death/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Hydrogen-Ion Concentration , Mice , Mice, Knockout , Phosphorylation/drug effects , Phosphorylation/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor-alpha/pharmacokinetics
2.
Mol Cell Biochem ; 464(1-2): 83-91, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31741130

ABSTRACT

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.


Subject(s)
Bifidobacterium , Intestinal Mucosa/metabolism , MAP Kinase Signaling System , Mucocutaneous Lymph Node Syndrome/prevention & control , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Caco-2 Cells , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Intestinal Mucosa/pathology , Mucocutaneous Lymph Node Syndrome/metabolism , Mucocutaneous Lymph Node Syndrome/pathology , Tumor Necrosis Factor-alpha/pharmacokinetics
3.
Blood ; 134(3): 252-262, 2019 07 18.
Article in English | MEDLINE | ID: mdl-31118164

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blood-Brain Barrier/drug effects , Central Nervous System Neoplasms/drug therapy , Lymphoma, Non-Hodgkin/drug therapy , Recombinant Fusion Proteins/pharmacokinetics , Tumor Necrosis Factor-alpha/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers , Blood-Brain Barrier/diagnostic imaging , CD13 Antigens/metabolism , Cell Membrane Permeability , Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/mortality , Cyclophosphamide/adverse effects , Cyclophosphamide/therapeutic use , Doxorubicin/adverse effects , Doxorubicin/therapeutic use , Female , Humans , Immunohistochemistry , Lymphoma, Non-Hodgkin/diagnosis , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/mortality , Male , Neuroimaging/methods , Prednisone/adverse effects , Prednisone/therapeutic use , Recombinant Fusion Proteins/administration & dosage , Research Design , Rituximab/adverse effects , Rituximab/therapeutic use , Tomography, Emission-Computed, Single-Photon , Treatment Outcome , Tumor Necrosis Factor-alpha/administration & dosage , Vincristine/adverse effects , Vincristine/therapeutic use
4.
J Biol Chem ; 293(52): 20062-20072, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30361438

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Multienzyme Complexes/metabolism , Tumor Necrosis Factor-alpha/pharmacokinetics , Ubiquitination/drug effects , Apoptosis/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , HEK293 Cells , HeLa Cells , Humans , Multienzyme Complexes/genetics , Ubiquitination/genetics
5.
Adv Healthc Mater ; 6(13)2017 Jul.
Article in English | MEDLINE | ID: mdl-28557249

ABSTRACT

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.


Subject(s)
Cell Cycle/drug effects , Drug Delivery Systems/methods , Nanoparticles , Neoplasms/drug therapy , Silicon Dioxide , Tumor Necrosis Factor-alpha , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms/metabolism , Porosity , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacokinetics , Silicon Dioxide/pharmacology , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/pharmacokinetics , Tumor Necrosis Factor-alpha/pharmacology
6.
Drug Metab Dispos ; 45(7): 798-806, 2017 07.
Article in English | MEDLINE | ID: mdl-28411279

ABSTRACT

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.


Subject(s)
Models, Biological , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/pharmacokinetics , Absorption, Physiological , Animals , Dose-Response Relationship, Drug , Haplorhini , Infusions, Subcutaneous , Injections, Intravenous , Injections, Subcutaneous , Male , Organ Specificity , Rats, Inbred Lew , Recombinant Proteins/blood , Species Specificity , Tissue Distribution , Tumor Necrosis Factor-alpha/blood
7.
Aliment Pharmacol Ther ; 45(10): 1329-1338, 2017 May.
Article in English | MEDLINE | ID: mdl-28318043

ABSTRACT

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.


Subject(s)
Adalimumab/metabolism , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Infliximab/pharmacokinetics , Maternal-Fetal Exchange , Adalimumab/pharmacology , Adalimumab/therapeutic use , Adolescent , Adult , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Crohn Disease/drug therapy , Crohn Disease/metabolism , Drug Monitoring , Female , Humans , Infliximab/therapeutic use , Maternal-Fetal Exchange/drug effects , Placenta/drug effects , Placenta/metabolism , Placental Circulation , Pregnancy , Tumor Necrosis Factor-alpha/pharmacokinetics , Tumor Necrosis Factor-alpha/therapeutic use , Young Adult
8.
Adv Mater ; 29(13)2017 Apr.
Article in English | MEDLINE | ID: mdl-28160337

ABSTRACT

A "relay drug delivery" system based on two distinct modules, which is composed of a signal transmission nanocarrier A (NCA ) that can specifically induce tumor blood vessel inflammation generation and an execution biomimetic nanocarrier B (NCB ) that can accumulate at the tumor site by receiving the broadcasting signals generated by NCA , is developed for amplifying active tumor targeting signal and enhancing antitumor therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Biomimetic Materials , Drug Delivery Systems , Nanostructures , Neoplasms/blood supply , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Biomimetic Materials/chemistry , Blood Vessels/drug effects , Blood Vessels/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Dextrans/chemistry , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice, Nude , Microtubules/enzymology , Microtubules/metabolism , Nanostructures/chemistry , Neoplasm Transplantation , Neoplasms/metabolism , Oligopeptides/chemistry , Paclitaxel/administration & dosage , Paclitaxel/pharmacokinetics , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/pharmacokinetics
9.
Biochemistry (Mosc) ; 81(10): 1188-1197, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27908243

ABSTRACT

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.


Subject(s)
Antioxidants/pharmacology , Capillary Permeability/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Plastoquinone/analogs & derivatives , Rhodamines/pharmacology , Tumor Necrosis Factor-alpha/pharmacokinetics , Antigens, CD/metabolism , Apoptosis/drug effects , Cadherins/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Humans , Mitochondria/metabolism , Plastoquinone/pharmacology
10.
Drug Deliv ; 23(6): 2044-57, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26599817

ABSTRACT

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.


Subject(s)
Antihypertensive Agents/chemistry , Drug Carriers/chemistry , Estrogens/chemistry , Genistein/chemistry , Genistein/pharmacology , Hypertension/drug therapy , Tumor Necrosis Factor-alpha/chemistry , Animals , Antihypertensive Agents/pharmacokinetics , Biological Availability , Chemistry, Pharmaceutical , Genistein/pharmacokinetics , Hypertension/metabolism , Rats , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacokinetics
11.
Sci Rep ; 5: 18363, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26678960

ABSTRACT

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.


Subject(s)
Chemistry, Pharmaceutical/methods , Tumor Necrosis Factor-alpha/chemistry , Amines/chemistry , Animals , Cell Line , Female , Half-Life , Mice , Mice, Inbred BALB C , Models, Molecular , Polyethylene Glycols/chemistry , Protein Structure, Tertiary , Succinimides/chemistry , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/pharmacokinetics
12.
J Rheumatol ; 42(12): 2376-82, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26568593

ABSTRACT

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.


Subject(s)
Spondylarthritis/diagnosis , Spondylarthritis/drug therapy , Spondylitis, Ankylosing/diagnosis , Spondylitis, Ankylosing/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adult , Age Factors , Cohort Studies , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Longitudinal Studies , Magnetic Resonance Imaging/methods , Male , Middle Aged , Pain Measurement , Retrospective Studies , Risk Assessment , Severity of Illness Index , Sex Factors , Treatment Outcome , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/pharmacokinetics
13.
Gastroenterology ; 149(2): 350-5.e2, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25917786

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Colitis, Ulcerative/drug therapy , Feces/chemistry , Intestinal Mucosa/drug effects , Adult , Antibodies, Monoclonal/administration & dosage , Colitis, Ulcerative/blood , Colitis, Ulcerative/pathology , Drug Monitoring/methods , Enzyme-Linked Immunosorbent Assay , Female , Humans , Infliximab , Intestinal Mucosa/pathology , Male , Middle Aged , Netherlands , Severity of Illness Index , Treatment Outcome , Tumor Necrosis Factor-alpha/pharmacokinetics , Tumor Necrosis Factor-alpha/therapeutic use
14.
Acta Biomater ; 12: 174-182, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25305513

ABSTRACT

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.


Subject(s)
Hyaluronic Acid/therapeutic use , Liver Cirrhosis/drug therapy , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacokinetics , Ligands , Male , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/pharmacokinetics
15.
Drug Dev Ind Pharm ; 41(3): 369-74, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24274583

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/administration & dosage , Hot Temperature/therapeutic use , Hydrogel, Polyethylene Glycol Dimethacrylate/administration & dosage , Injections, Intralesional/methods , Tumor Necrosis Factor-alpha/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacokinetics , Male , Mice , Mice, Inbred ICR , Tumor Necrosis Factor-alpha/pharmacokinetics
16.
J. physiol. biochem ; 70(4): 971-980, dic. 2014.
Article in English | IBECS | ID: ibc-131430

ABSTRACT

Tumor necrosis factor-alfa (TNF-alfa), a pro-apoptotic cytokine, is involved in vascular hyperpermeability, tissue edema, and inflammation. We hypothesized that TNF-alfa induces microvascular hyperpermeability through the mitochondria-mediated intrinsic apoptotic signaling pathway. Rat lung microvascular endothelial cells grown on Transwell inserts, chamber slides, or dishes were treated with recombinant TNF-alfa (10 ng/ml) in the presence or absence of a caspase-3 inhibitor, Z-DEVD-FMK (100 μM). Fluorescein isothiocyanate (FITC)-albumin (5 mg/ml) was used as a marker of monolayer permeability. Mitochondrial reactive oxygen species (ROS) was determined using dihydrorhodamine 123 and mitochondrial transmembrane potential using JC-1. The adherens junction integrity and actin cytoskeletal organization were studied using β-catenin immunofluorescence and rhodamine phalloidin, respectively. Caspase-3 activity was measured fluorometrically. The pretreatment with Z-DEVD-FMK (100 μM) attenuated TNF-alfa-induced (10 ng/ml) disruption of the adherens junctions, actin stress fiber formation, increased caspase-3 activity, and monolayer hyperpermeability (p < 0.05). TNF-alfa (10 ng/ml) treatment resulted in increased mitochondrial ROS formation and decreased mitochondrial transmembrane potential. Intrinsic apoptotic signaling-mediated caspase-3 activation plays an important role in regulating TNF-α-induced endothelial cell hyperpermeability


Subject(s)
Animals , Rats , Tumor Necrosis Factor-alpha/pharmacokinetics , Capillary Permeability , Mitochondria , TNF-Related Apoptosis-Inducing Ligand , Adherens Junctions , Cadherins , beta Catenin , Caspase 3 , Inflammation/physiopathology
17.
J. physiol. biochem ; 70(3): 769-779, sept. 2014.
Article in English | IBECS | ID: ibc-127321

ABSTRACT

An elevated level of tumor necrosis factor (TNF)-α is implicated in several cardiovascular diseases including heart failure. Numerous reports have demonstrated that TNF- alfa activates nuclear factor (NF)-kappaB, resulting in the upregulation of several genes that regulate inflammation, proliferation, and apoptosis of cardiomyocytes. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, a major source of reactive oxygen species (ROS), is also activated by TNF- alfa and plays a crucial role in redox-sensitive signaling pathways. The present study investigated whether NADPH oxidase mediates TNF-alfa-induced NF-kappaB activation and NF-kappaB-mediated gene expression. Human cardiomyocytes were treated with recombinant TNF- alfa with or without pretreatment with diphenyleneiodonium (DPI) and apocynin, inhibitors of NADPH oxidase. TNF-α-induced ROS production was measured using 5-(and-6)-chloromethyl-2', 7'-dichlorodihydrofluorescein diacetate assay. TNF- alfa-induced NF-kappaB activation was also examined using immunoblot; NF-kappaB binding to its binding motif was determined using a Cignal reporter luciferase assay and an electrophoretic mobility shift assay. TNF- alfa -induced upregulation of interleukin (IL)-1β and vascular cell adhesion molecule (VCAM)-1 was investigated using real-time PCR and immunoblot. TNF- alfa -induced ROS production in cardiomyocytes was mediated by NADPH oxidase. Phosphorylation of IKK- alfa /β and p65, degradation of IkappaBalfa, binding of NF-kappaB to its binding motif, and upregulation of IL-1β and VCAM-1 induced by TNF- alfa were significantly attenuated by treatment with DPI and apocynin. Collectively, these findings demonstrate that NADPH oxidase plays a role in regulation of TNF- alfa -induced NF-kappaB activation and upregulation of proinflammatory cytokines, IL-1β and VCAM-1, in human cardiomyocytes


Subject(s)
Humans , Tumor Necrosis Factor-alpha/pharmacokinetics , Receptor Activator of Nuclear Factor-kappa B , Myocytes, Cardiac , NADPH Oxidases/pharmacokinetics , MAP Kinase Signaling System , Inflammation Mediators , Inflammation/physiopathology
18.
J Parkinsons Dis ; 4(3): 349-60, 2014.
Article in English | MEDLINE | ID: mdl-25061061

ABSTRACT

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.


Subject(s)
Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/pathology , Substantia Nigra/drug effects , Substantia Nigra/pathology , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Blood-Brain Barrier , Cell Count , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Encephalitis/prevention & control , Gliosis/prevention & control , Male , Oxidopamine , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/pharmacokinetics
19.
J Biotechnol ; 186: 1-12, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-24992210

ABSTRACT

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.


Subject(s)
Antibodies, Bispecific/therapeutic use , Arthritis, Experimental/drug therapy , Fibronectins/therapeutic use , Protein Engineering/methods , Tumor Necrosis Factor-alpha/therapeutic use , Amino Acid Sequence , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Bispecific/pharmacokinetics , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Base Sequence , Cell Line, Tumor , Cytokines/analysis , Cytokines/metabolism , Disease Progression , Fibronectins/chemistry , Fibronectins/genetics , Fibronectins/pharmacokinetics , Foot/pathology , Mice , Molecular Sequence Data , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Tissue Distribution , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/pharmacokinetics
20.
Eur J Pharmacol ; 740: 722-32, 2014 Oct 05.
Article in English | MEDLINE | ID: mdl-24929054

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Membrane Glycoproteins , Recombinant Fusion Proteins , Tumor Necrosis Factor-alpha , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Clinical Trials, Phase I as Topic , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Hepatocytes/drug effects , Humans , Macaca fascicularis , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/pharmacokinetics , Membrane Glycoproteins/pharmacology , Membrane Glycoproteins/therapeutic use , Mice, Inbred BALB C , Mice, Nude , Neoplasms/drug therapy , Neoplasms/pathology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , TNF-Related Apoptosis-Inducing Ligand/pharmacokinetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Tumor Burden/drug effects , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/pharmacokinetics , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/therapeutic use , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL