Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
1.
Protein Expr Purif ; 162: 32-37, 2019 10.
Article in English | MEDLINE | ID: mdl-31100416

ABSTRACT

In this study, canine IFNγ was fused by a flexible linker with canine serum albumin to construct the fusion protein IFNγ-CSA for the purpose to design a long-acting canine IFNγ. The fusion protein was successfully expressed in baculovirus-infected Sf9 insect cells and was purified by salting-out and ion exchange chromatography. The IFNγ-CSA fusion possessed potent anti-viral assay against vesicular stomatitis virus in cultured cells. IFNγ-CSA was also stable at 37 °C up to 72 h compared with 8 h for IFNγ alone. In vivo pharmacokinetics demonstrated a significantly longer half-life for IFNγ-CSA (15.42 h) than for canine reIFNγ (1.51 h) in KM mice. These results indicate that IFNγ-CSA expression in the baculovirus system was successful and provide a promising long-acting cytokine for veterinary clinical applications.


Subject(s)
Baculoviridae/genetics , Interferon-gamma/genetics , Serum Albumin/genetics , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacokinetics , Baculoviridae/metabolism , Dogs , Female , Gene Expression , Interferon-gamma/metabolism , Interferon-gamma/pharmacokinetics , Mice , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacokinetics , Serum Albumin/metabolism , Serum Albumin/pharmacokinetics , Sf9 Cells , Spodoptera , Vesicular stomatitis Indiana virus/drug effects
2.
Cytokine ; 97: 25-37, 2017 09.
Article in English | MEDLINE | ID: mdl-28558308

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in young children and is further associated with increased healthcare utilization and cost of care in the first years of life. Severe RSV disease during infancy has also been linked to the later development of allergic asthma, yet there remains no licensed RSV vaccine or effective treatment. Pre-clinical and clinical studies have shown that disease severity and development of allergic asthma are associated with differences in cytokine production. As a result, stimulation of the innate host immune response with immune potentiators is gaining attention for their prospective application in populations with limited immune responses to antigenic stimuli or against pathogens for which vaccines do not exist. Specifically, macrophage-activating cytokines such as interferon gamma (IFNγ) and granulocyte colony-stimulating factor (GM-CSF) are commercially available immune potentiators used to prevent infections in patients with chronic granulomatous disease and febrile neutropenia, respectively. Moreover, an increasing number of reports describe the protective function of IFNγ and GM-CSF as vaccine adjuvants. Although a positive correlation between cytokine production and age has previously been reported, little is known about age-dependent cytokine metabolism or immune activating responses in infant compared to adult lungs. Here we use a non-compartmental pharmacokinetic model in naïve and RSV-infected infant and adult BALB/c mice to determine the effect of age on IFNγ and GM-CSF elimination and innate cell activation following intranasal delivery.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Immunity, Innate/drug effects , Interferon-gamma/administration & dosage , Respiratory Syncytial Virus Infections/immunology , Administration, Intranasal , Age Factors , Animals , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/blood , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacokinetics , Interferon-gamma/blood , Interferon-gamma/immunology , Interferon-gamma/pharmacokinetics , Kinetics , Lung/drug effects , Lung/immunology , Lung/virology , Macrophage Activation , Mice , Mice, Inbred BALB C , Prospective Studies , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/physiology
3.
Respir Res ; 16: 122, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26438053

ABSTRACT

BACKGROUND: Poor interferon gamma (IFNγ) production during respiratory syncytial virus (RSV) is associated with prolonged viral clearance and increased disease severity in neonatal mice and humans. We previously showed that intra-nasal delivery of IFNγ significantly enhances RSV clearance from neonatal lungs prior to observed T-lymphocyte recruitment or activation, suggesting an innate immune mechanism of viral clearance. We further showed that alveolar macrophages dominate the RSV-infected neonatal airways relative to adults, consistent with human neonatal autopsy data. Therefore, the goal of this work was to determine the role of neonatal alveolar macrophages in IFNγ-mediated RSV clearance. METHODS: Clodronate liposomes, flow cytometry, viral plaque assays, and histology were used to examine the role of alveolar macrophages (AMs) and the effects of intra-nasal IFNγ in RSV infected neonatal Balb/c mice. The functional outcomes of AM depletion were determined quantitatively by viral titers using plaque assay. Illness was assessed by measuring reduced weight gain. RESULTS: AM activation during RSV infection was age-dependent and correlated tightly with IFNγ exposure. Higher doses of IFNγ more efficiently stimulated AM activation and expedited RSV clearance without significantly affecting weight gain. The presence of AMs were independently associated with improved RSV clearance, whereas AM depletion but not IFNγ exposure, significantly impaired weight gain in RSV-infected neonates. CONCLUSION: We show here for the first time, that IFNγ is critical for neonatal RSV clearance and that it depends, in part, on alveolar macrophages (AMs) for efficient viral clearing effects. Early reductions in viral burden are likely to have profound short- and long-term immune effects in the vulnerable post-natally developing lung environment. Studies are ongoing to elucidate the pathologic effects associated with early versus delayed RSV clearance in developing neonatal airways.


Subject(s)
Antiviral Agents/administration & dosage , Interferon-gamma/administration & dosage , Macrophage Activation/drug effects , Macrophages, Alveolar/drug effects , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects , Administration, Intranasal , Age Factors , Animals , Animals, Newborn , Antiviral Agents/pharmacokinetics , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Interferon-gamma/pharmacokinetics , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/virology , Male , Mice, Inbred BALB C , Mice, Knockout , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , Time Factors , Viral Load , Weight Gain , Interferon gamma Receptor
4.
Nanotechnology ; 23(26): 265101, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22700686

ABSTRACT

No satisfactory anti-fibrotic therapies have yet been applied clinically. One of the main reasons is the inability to specifically target the responsible cells to produce an available drug concentration and the side-effects. Exploiting the key role of the activated hepatic stellate cells (HSCs) in both hepatic fibrogenesis and over-expression of platelet-derived growth factor receptor- (PDGFR- ), we constructed targeted sterically stable liposomes (SSLs) modified by a cyclic peptide (pPB) with affinity for the PDGFR- to deliver interferon (IFN)- to HSCs. The pPB-SSL-IFN- showed satisfactory size distribution. In vitro pPB-SSL could be taken up by activated HSCs. The study of tissue distribution via living-body animal imaging showed that the pPB-SSL-IFN- mostly accumulated in the liver until 24 h. Furthermore, the pPB-SSL-IFN- showed more significant remission of hepatic fibrosis. In vivo the histological Ishak stage, the semiquantitative score for collagen in fibrotic liver and the serum levels of collagen type IV-C in fibrotic rats treated with pPB-SSL-IFN- were less than those treated with SSL-IFN- , IFN- and the control group. In vitro pPB-SSL-IFN- was also more effective in suppressing activated HSC proliferation and inducing apoptosis of activated HSCs. Thus the data suggest that pPB-SSL-IFN- might be a more effective anti-fibrotic agent and a new opportunity for clinical therapy of hepatic fibrosis.


Subject(s)
Hepatic Stellate Cells/metabolism , Interferon-gamma/administration & dosage , Liposomes/administration & dosage , Liver Cirrhosis/drug therapy , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Hepatic Stellate Cells/drug effects , Histocytochemistry , Interferon-gamma/chemistry , Interferon-gamma/pharmacokinetics , Liposomes/chemistry , Liposomes/pharmacokinetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacokinetics , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Rats , Rats, Sprague-Dawley , Spectrometry, Fluorescence , Tissue Distribution , Whole Body Imaging
5.
Life Sci ; 264: 118605, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33096119

ABSTRACT

The purpose of this study was to prepare non-PEGylated (HSPC/DSPG/Chol, LIPF1) and PEGylated (HSPC/DSPG/Chol/mPEG2000-DSPE, LIPF2) liposomal formulations containing Interferon-gamma (IFN-γ) and evaluation their effects on macrophages and their antitumor properties. The results showed that the size of liposomal formulations LIP-F1 and LIP-F2 was 120 and 135 nm, respectively. The encapsulation efficiencies of LIP-F1 and LIP-F2 were 52.79% and 49.2%, respectively. Nitric Oxide Synthase (INOS) and arginase assays showed an increase in nitric oxide (NO) level and a reduction in arginase level after the treatment of M2 phenotype macrophage cell line with IFN-γ liposomes. The biodistribution study illustrated the amplitude of iodinated-IFN-γ liposomal formulations in the tumor site, the circulation time and tumor accumulation of LIP-F2 was significantly more than LIPF1. As a result, PEGylated liposomes containing IFN-γ induced significant antitumor responses due to the increased delivery of the cargo to the immune cells and induction of antitumor immune responses.


Subject(s)
Colonic Neoplasms/drug therapy , Disease Models, Animal , Immunotherapy/methods , Interferon-gamma/administration & dosage , Nanoparticles/administration & dosage , Animals , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Female , Interferon-gamma/immunology , Interferon-gamma/pharmacokinetics , Liposomes , Mice , Mice, Inbred BALB C , Nanoparticles/metabolism , Random Allocation , Tissue Distribution/drug effects , Tissue Distribution/physiology
6.
Med Mycol ; 48(3): 532-6, 2010 May.
Article in English | MEDLINE | ID: mdl-19835489

ABSTRACT

We present a case of relapsing cryptococcal meningitis unresponsive to standard therapy. Voriconazole induction, including the utilization of voriconazole therapeutic drug monitoring in both serum and CSF, with transition to voriconazole plus interferon-gamma (IFN-gamma) was successfully used in a patient receiving antiretroviral therapy with abacavir/lamivudine and lopinavir/ritonavir. Initial voriconazole levels at standard doses of 4 mg/kg twice daily intravenously were low when co-administered with lopinavir/ritonavir but increased to recommended therapeutic levels with an increase of the voriconazole dose to 7 mg/kg twice daily. This case highlights the utility of voriconazole therapeutic drug monitoring when prescribed concurrently with a ritonavir boosted protease inhibitor and the potential role of combination therapy with IFN-G for refractory cryptococcal meningitis.


Subject(s)
Antifungal Agents/pharmacokinetics , HIV Infections/complications , Interferon-gamma/pharmacokinetics , Meningitis, Cryptococcal/drug therapy , Pyrimidines/pharmacokinetics , Triazoles/pharmacokinetics , Anti-HIV Agents/therapeutic use , Antifungal Agents/administration & dosage , Antifungal Agents/therapeutic use , Cerebrospinal Fluid/chemistry , Dideoxynucleosides/therapeutic use , Drug Monitoring , HIV Infections/drug therapy , Humans , Interferon-gamma/therapeutic use , Lamivudine/therapeutic use , Male , Pyrimidines/administration & dosage , Pyrimidines/therapeutic use , Ritonavir/therapeutic use , Serum/chemistry , Triazoles/administration & dosage , Triazoles/therapeutic use , Voriconazole , Young Adult
7.
J Aerosol Med Pulm Drug Deliv ; 33(2): 108-115, 2020 04.
Article in English | MEDLINE | ID: mdl-31855492

ABSTRACT

Background: In volunteers with idiopathic pulmonary fibrosis (IPF), inhaled Interferon-γ (IFN-γ) is safe and may improve pulmonary function. However, coughing, associated with upper airway deposition, is often reported. To address this problem, a small-particle, breath-enhanced jet nebulizer (i-NEB Mini; InspiRx, Inc., Somerset, NJ) was developed. Using gamma scintigraphy, this device was tested in healthy individuals and subjects with IPF to determine efficiency and regional deposition in lung and airways. Methods: Four healthy individuals and nine subjects with IPF were enrolled. The nebulizer was filled with 2 mL of saline with 99m Tc bound to diethylenetriaminepentaacetic acid (DTPA) powered continuously with 3.4 L/min of compressed air. Mass median aerodynamic diameter (MMAD) was measured by cascade impactor. To maximize deposition in alveoli, inspiratory flow was limited by an inspiratory resistance incorporated into the nebulizer, resulting in a deep inspiration ∼6 seconds. The treatment was run to completion (10 minutes), and each subject underwent deposition imaging. Mass balance and regions of interest determined upper airway (measured by calibrated stomach activity) and regional lung deposition as a percent of pretreatment nebulizer charge. Results: Subjects tolerated the device with no complaints. MMAD (mean [geometric standard deviation]) = 1.04 [1.92] µm. Lung deposition (mean ± standard error, % nebulizer charge) in healthy subjects was 26.2% ± 1.83 and in IPF individuals 23.4% ± 1.60 (p = 0.414). Upper airway deposition was 1.4% ± 0.83 and 2.3% ± 0.48, respectively (p = 0.351), and 20.1% was lost during expiration. Central/Peripheral ratios were consistent in both groups, showing high peripheral deposition (1.32 ± 0.050, vs. 1.28 ± 0.046, p = 0.912). Conclusion: The i-NEB Mini jet nebulizer with breath enhancement produced small particles, resulting in minimal upper airway deposition. Using slow and deep breathing, more than half of the emitted dose deposited in the peripheral lung in normal subjects and individuals with IPF. These data indicate that, for future clinical trials, controlled lung doses of small particles, designed to avoid coughing, are possible even in subjects with advanced disease.


Subject(s)
Idiopathic Pulmonary Fibrosis/drug therapy , Interferon-gamma/administration & dosage , Lung/metabolism , Nebulizers and Vaporizers , Administration, Inhalation , Case-Control Studies , Cough/etiology , Equipment Design , Humans , Interferon-gamma/adverse effects , Interferon-gamma/pharmacokinetics , Particle Size , Tissue Distribution
8.
Int J Cancer ; 125(1): 115-23, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19384924

ABSTRACT

Immunotherapeutic targeting of G250/Carbonic anhydrase IX (CA-IX) represents a promising strategy for treatment of renal cell carcinoma (RCC). The well characterized human-mouse chimeric G250 (cG250) antibody has been shown in human studies to specifically enrich in CA-IX positive tumors and was chosen as a carrier for site specific delivery of TNF in form of our IgG-TNF-fusion protein (cG250-TNF) to RCC xenografts. Genetically engineered TNF constructs were designed as CH2/CH3 truncated cG250-TNF fusion proteins and eucariotic expression was optimized under serum-free conditions. In-vitro characterization of cG250-TNF comprised biochemical analysis and bioactivity assays, alone and in combination with Interferon-gamma (IFNgamma). Biodistribution data on radiolabeled [(125)J] cG250-TNF and antitumor activity of cG250-TNF, alone and in combination with IFNgamma, were measured on RCC xenografts in BALB/c nu/nu mice. Combined administration of cG250-TNF and IFNgamma caused synergistic biological effects that represent key mechanisms displaying antitumor responses. Biodistribution studies demonstrated specific accumulation and retention of cG250-TNF at CA-IX-positive RCC resulting in growth inhibition of RCC and improved progression free survival and overall survival. Antitumor activity induced by targeted TNF-based constructs could be enhanced by coadministration of low doses of nontargeted IFNgamma without significant increase in side effects. Administration of cG250-TNF and IFNgamma resulted in significant synergistic tumoricidal activity. Considering the poor outcome of renal cancer patients with advanced disease, cG250-TNF-based immunotherapeutic approaches warrant clinical evaluation.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Carcinoma, Renal Cell/therapy , Interferon-gamma/therapeutic use , Kidney Neoplasms/therapy , Recombinant Fusion Proteins/therapeutic use , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Antibodies, Monoclonal/pharmacokinetics , Antigens, Neoplasm/immunology , Cancer Vaccines/pharmacokinetics , Cancer Vaccines/therapeutic use , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Cells, Cultured , Drug Synergism , Drug Therapy, Combination , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Flow Cytometry , Humans , Immunoglobulin G/therapeutic use , Interferon-gamma/pharmacokinetics , Iodine Radioisotopes/pharmacokinetics , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Proteins , Tissue Distribution , Tumor Necrosis Factor-alpha/pharmacokinetics , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism , Xenograft Model Antitumor Assays
9.
Science ; 269(5225): 850-3, 1995 Aug 11.
Article in English | MEDLINE | ID: mdl-7638603

ABSTRACT

Transdermal drug delivery offers a potential method of drug administration. However, its application has been limited to a few low molecular weight compounds because of the extremely low permeability of human skin. Low-frequency ultrasound was shown to increase the permeability of human skin to many drugs, including high molecular weight proteins, by several orders of magnitude, thus making transdermal administration of these molecules potentially feasible. It was possible to deliver and control therapeutic doses of proteins such as insulin, interferon gamma, and erythropoeitin across human skin. Low-frequency ultrasound is thus a potential noninvasive substitute for traditional methods of drug delivery, such as injections.


Subject(s)
Administration, Cutaneous , Epidermis/metabolism , Insulin/administration & dosage , Phonophoresis , Proteins/administration & dosage , Blood Glucose/analysis , Diabetes Mellitus/drug therapy , Erythropoietin/administration & dosage , Erythropoietin/pharmacokinetics , Humans , Insulin/pharmacokinetics , Interferon-gamma/administration & dosage , Interferon-gamma/pharmacokinetics , Permeability , Proteins/pharmacokinetics , Skin Absorption , Transducers
10.
Int J Pharm ; 558: 404-412, 2019 Mar 10.
Article in English | MEDLINE | ID: mdl-30639219

ABSTRACT

Interferon (IFN)-γ plays an important role in antiviral, anti-proliferative, immunomodulatory and pro-inflammatory activities. However, the short therapeutic half-life of IFN-γ lessens its efficacy. Albumin fusion strategy is one of the most effective ways to improve the pharmacokinetic properties of cytokines. In this study, N- and C-terminal canine albumin fusions with canine IFN-γ were expressed in the baculovirus expression system. The fusion proteins stimulated Stat1 phosphorylation at levels similar to that of the recombinant IFN. The antiviral, anti-proliferative and promote apoptosis activity of CSA-IFN-γ was lower than IFN-γ-CSA and both were less than that of recombinant IFN-γ. In vivo pharmacokinetics demonstrated a significantly longer half-life for CSA-IFN-γ (21.73 h) than for IFN-γ-CSA (6.51 h) and canine reIFN-γ (2.22 h) in Wistar rats. CSA-IFN-γ was also more effective than IFN-γ-CSA and canine reIFN-γ at inhibiting growth of canine renal malignant histiocytosis in nude mice. Our results indicated that a canine serum albumin fusion at the N-terminus of IFN-γ prolongs its half-life and improves its in vivo antitumor activity.


Subject(s)
Antineoplastic Agents/administration & dosage , Interferon-gamma/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Serum Albumin/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Proliferation/drug effects , Female , Interferon-gamma/chemistry , Interferon-gamma/pharmacokinetics , Mice, Inbred BALB C , Mice, Nude , Neoplasms/drug therapy , Rats, Wistar , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Serum Albumin/chemistry , Serum Albumin/pharmacokinetics
11.
Inflamm Bowel Dis ; 14(3): 303-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18050297

ABSTRACT

BACKGROUND: The intestinal epithelium is a single layer of polarized cells and is the primary barrier separating foreign antigen and underlying lymphoid tissue. IFNgamma alters epithelial barrier function during inflammation by disrupting tight cell junctions and facilitating the paracellular transport of luminal antigens. The aim of this work was to determine whether Campylobacter infection of cells exposed to IFNgamma would lead to greater disruption of cell monolayers and hence increased bacterial translocation. METHODS: Monolayers were polarized on Transwell polycarbonate membranes for 14 days and then cultured in the presence or absence of 100 U/mL IFNgamma. Campylobacter was added to the apical side of the monolayer at an MOI of 30. Transepithelial electrical resistance (TEER) was recorded and bacteria in the basal well counted every 2 hours. Cells were stained for occludin, actin, and nuclear DNA, and cell viability determined by measurement of apoptosis. RESULTS: In the presence of IFNgamma, TEER dropped significantly after 18 hours, indicating a reduction in barrier function. A further significant decrease was seen in the presence of both IFNgamma and Campylobacter, indicating a synergistic effect, and cellular morphology and viability were affected. Bacterial translocation across the monolayer was also significantly greater in the presence of IFNgamma. CONCLUSIONS: These combined effects indicate that Campylobacter infection concomitant with intestinal inflammation would result in a rapid and dramatic loss of epithelial barrier integrity, which may be a key event in the pathogenesis of Campylobacter-mediated colitis and the development of bloody diarrhea.


Subject(s)
Bacterial Translocation/physiology , Campylobacter jejuni/physiology , Cell Membrane Permeability/drug effects , Interferon-gamma/pharmacokinetics , Intestinal Mucosa/metabolism , Actins/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Bacterial Translocation/drug effects , Caco-2 Cells , Campylobacter Infections/metabolism , Campylobacter Infections/microbiology , Campylobacter Infections/pathology , Epithelium/metabolism , Epithelium/microbiology , Epithelium/pathology , Humans , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Membrane Potentials/drug effects , Membrane Proteins/metabolism , Microscopy, Fluorescence , Occludin
12.
Semin Oncol ; 45(1-2): 27-33, 2018 01.
Article in English | MEDLINE | ID: mdl-30318081

ABSTRACT

The rational combination of recombinant IFN-α2b and IFN-γ resulted in a new formulation of interferons (HeberFERON) with improved pharmacodynamics. In basal cell carcinomas HeberFERON produces a more rapid antitumor effect and results in a larger number of complete responses. In patients with glioblastoma multiforme, the administration of HeberFERON after surgery and radiotherapy results in an estimated overall survival of 19 months. Patients with stage III or IV renal cell carcinoma also appear to benefit from the intravenous administration of HeberFERON, with prolongation of survival and good quality of live. HeberFERON offers a promising alternative formulation of interferons for the treatment of cancer with a very favorable safety profile.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Interferon-alpha/therapeutic use , Interferon-gamma/therapeutic use , Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic , Humans , Interferon alpha-2 , Interferon-alpha/administration & dosage , Interferon-alpha/pharmacokinetics , Interferon-gamma/administration & dosage , Interferon-gamma/pharmacokinetics , Neoplasms/genetics , Neoplasms/metabolism , Proteome/metabolism , Quality of Life , Survival Analysis , Treatment Outcome
13.
Protein Eng Des Sel ; 31(9): 355-360, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30496575

ABSTRACT

The previously generated recombinant human (rh) interferon (IFN)-λ1 protein has a short half-life, and this feature makes it challenging to conduct studies on potential clinical applications for rhIFN-λ1. In an attempt to overcome this difficulty, we constructed a 'long-life' version of rhIFN-λ1. This modified rhIFN-λ1, named rhIFN-λ1-CTPON, has a human chorionic gonadotropin ß subunit carboxyl-terminal peptide (CTP) and an N-glycosylation sequence linked to its C-terminus. We confirmed the sequence of rhIFN-λ1-CTPON by mass spectrometry and then measured its biological activities. The results show that rhIFN-λ1-CTPON had antiviral activity and anti-proliferation activity in vitro that were similar to those of rhIFN-λ1 and that it similarly promoted natural killer cell cytotoxicity. Notably, the in vivo half-life of rhIFN-λ1-CTPON was determined to be 3-fold higher than that of rhIFN-λ1. We also assessed the anti-hepatitis B virus activity of rhIFN-λ1-CTPON; it was able to inhibit the production of the antigens HBs-Ag and HBe-Ag and induce antiviral gene expression. In conclusion, rhIFN-λ1-CTPON has a longer half-life than rhIFN-λ1 and has similar biological activities, so rhIFN-λ1-CTPON is an appropriate substitute for rhIFN-λ1 in the further study of potential clinical applications for rhIFN- λ1.


Subject(s)
Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Chorionic Gonadotropin, beta Subunit, Human/genetics , Gene Expression/drug effects , Genes, Viral/genetics , Humans , Interferon-gamma/genetics , Interferon-gamma/pharmacokinetics , Male , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics
14.
Infect Immun ; 75(12): 5615-26, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17893127

ABSTRACT

Vascular endothelium is an exposed target in systemic endovascular Staphylococcus aureus infections. We reported earlier that the proinflammatory and procoagulant activities of primary human umbilical vein endothelial cells (ECs) after binding and ingestion of S. aureus organisms provide the cells effective means for leukocyte-mediated bacterial elimination. Expanding on this, we now show that these ECs exhibit a modest intrinsic capacity for eliminating intracellular S. aureus that was influenced by cytokines relevant to S. aureus infections. Using various EC infection assays, we showed that gamma interferon (IFN-gamma), applied to cultures of ECs prior to or after infection with S. aureus, markedly reduced the level of infection, illustrated by lower percentages of S. aureus-infected ECs and less intracellular bacteria per infected cell. IFN-gamma-activated ECs had unaltered abilities to bind S. aureus and processed ingested bacteria by a seemingly conventional phagocytic pathway. IFN-gamma treatment rescued EC monolayers from severe injury by virulent clinical S. aureus strains or excessive bacterial numbers. Mechanistically, IFN-gamma controls S. aureus infection via IFN-gamma receptor, most likely through stimulation of intrinsic endothelial antibacterial mechanisms but independent of processes that deprive bacteria of intracellular L-tryptophan or iron. The antibacterial activity of IFN-gamma-stimulated ECs coincided with sustained or slightly elevated endothelial proinflammatory responses that supported monocyte recruitment. In conclusion, we identify IFN-gamma as a potent regulatory Th1 cytokine possessing exclusive abilities to augment intrinsic antistaphylococcal effector mechanisms in human ECs without ablating the S. aureus-induced proinflammatory EC responses and, as such, coordinating a protective efficacy of ECs against blood-borne S. aureus infection.


Subject(s)
Endothelial Cells/immunology , Interferon-gamma/pharmacology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , Cattle , Dose-Response Relationship, Immunologic , Endocytosis/immunology , Endocytosis/physiology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Ferric Compounds/pharmacology , Humans , Inflammation/immunology , Interferon-gamma/immunology , Interferon-gamma/pharmacokinetics , Iron/metabolism , Recombinant Proteins , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/pathogenicity , Tryptophan/metabolism , Tryptophan/pharmacology
15.
Curr Opin Investig Drugs ; 8(6): 493-8, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17621880

ABSTRACT

Transgene SA is developing TG-1042, a replication-deficient adenovirus type 5 that carries the IFNgamma gene, for the potential treatment of cutaneous T-cell and B-cell lymphoma (CTCL and CBCL, respectively). A phase I/II clinical trial in CTCL and CBCL was recently completed, and in November 2006 Transgene initiated a phase II clinical trial in CBCL.


Subject(s)
Adenoviridae/genetics , Antineoplastic Agents/therapeutic use , Genetic Therapy , Interferon-gamma/genetics , Interferon-gamma/therapeutic use , Lymphoma, T-Cell, Cutaneous/drug therapy , Skin Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Humans , Interferon-gamma/chemical synthesis , Interferon-gamma/pharmacokinetics , Interferon-gamma/pharmacology , Lymphoma, T-Cell, Cutaneous/pathology , Skin Neoplasms/pathology
16.
Int J Clin Pharmacol Ther ; 45(6): 307-18, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17595888

ABSTRACT

OBJECTIVE: Application-site disorders are well-known adverse events (AEs) associated with subcutaneous (s.c.) injection. With high-dose, high-frequency interferon (IFN)-beta1a (Rebif) these AEs are generally mild but may lead to the discontinuation of some patients. The objective of this study was to compare the safety, tolerability, and the pharmacokinetic (PK) and pharmacodynamic (PD) profiles of two new formulations of Rebif (Rebif New Formulation: RNF1 and RNF2) with the current formulation (hereafter referred to as R) and placebo. METHODS: In this double-blind, placebo-controlled, parallel-group, Phase I study, healthy volunteers of both sexes were randomized 1:1:1:1 to receive a single 0.5 ml s.c. dose of RNF1, RNF2, R or placebo (normal saline). The three active treatments contained 44 microg IFN-beta1a. During the 24-hour post-dose period, safety and tolerability assessments were conducted and blood samples were taken at regular intervals for PK and PD analyses. Pain intensity on injection was measured using the short-form McGill questionnaire and a 100 mm visual analogue scale (VAS). Further safety assessments were performed and blood samples taken at 24-hour intervals until Day 7 post-dose, with a final post-study visit 10- 14 days after dosing. RESULTS: A total of 48 subjects (22 men, 26 women) were recruited and allocated equally to each treatment (12 subjects per group). AEs were reported by 10 subjects in each active treatment group and by 3 subjects in the placebo group. All AEs were consistent with the known safety profile of R. The number of treatment-emergent AEs was lower in the RNF2 group than the RNF 1 or R groups (21, 31 and 33 events, respectively). Redness at the injection site was mostly mild and occurred in fewer subjects in the RNF2 group (n = 3) than the RNF 1 or R groups (n = 7 and n = 4, respectively). Injection site pain was reported by 1 subject in the RNF2 group, compared with 4, 6 and 3 subjects, respectively, in the RNF1, R and placebo groups. The worst pain intensity, as measured by VAS, was lower in the RNF2 and RNFI groups than either the R or placebo groups. There was considerable intersubject variability in the PK and PD profiles of the three formulations of IFN-beta1a. Nevertheless, the PK and PD characteristics of RNF2 were similar to those of R. CONCLUSIONS: The results from this study suggest that RNF2 may offer improved tolerability compared with the current formulation of R, but retains comparable pharmacokinetic and pharmacodynamic characteristics.


Subject(s)
Interferon-gamma/pharmacology , Interferon-gamma/pharmacokinetics , Adolescent , Adult , Area Under Curve , Chemistry, Pharmaceutical , Double-Blind Method , Female , Half-Life , Humans , Hydrogen-Ion Concentration , Injections, Subcutaneous , Interferon-gamma/adverse effects , Male , Middle Aged , Neopterin/blood , Pain/chemically induced , Pain/epidemiology , Pain Measurement/drug effects , Recombinant Proteins , Surveys and Questionnaires , Treatment Outcome , beta 2-Microglobulin/blood
17.
J Interferon Cytokine Res ; 26(7): 473-83, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16800786

ABSTRACT

The standard of care for chronic hepatitis C, pegylated interferon-alpha (IFN-alpha) and ribavirin (RBV), causes a sustained virologic response (SVR) in approximately 50% of patients. SVR is correlated with innate and adaptive immune system responses, such as natural killer (NK) cell activation, production of IFN-alpha from immature plasmocytoid dendritic cells (pDC), and polarization of CD4(+) cells to a T helper 1 (Th1) cell phenotype. To examine how these immunologic responses vary with currently available regimens for chronic hepatitis C, cell populations purified from human peripheral blood mononuclear cells (PBMC) were treated with the clinically available combinations of pegylated IFN-alpha2b (PEG-IFN-alpha2b) + RBV, IFN-alphacon1 + RBV, or IFN- alphacon1 + IFN-gamma1b, and activation of cellular immune system components was monitored. The magnitude of NK cell activation depended on regimen, with IFN-alphacon1 + IFN-gamma1b > IFN-alphacon1 + RBV > PEG-IFN- alphaa2b + RBV. The maximum human serum concentrations of IFN-alphacon1 + IFN-gamma1b saturated NK cell activation, whereas the maximum human serum concentrations of IFN-alphacon1 + RBV or PEG-IFN-alpha2b + RBV did not. IFN-gamma1b also enhanced the production of IFN-alpha from immature pDCs, which are the dominant source of IFN-alpha upon viral infection. The rank order for induction of Th1 cell phenotype and repression of Th2 cell phenotype by the cocktails described was identical to that observed for NK cell activation. Additionally, IFN- gamma1b suppressed the ability of the hepatitis C virus (HCV) NS4 protein to enhance monocyte secretion of interleukin- 10 (IL-10), a cytokine whose expression level is correlated with viral persistence. These results suggest that addition of IFN-gamma1b to HCV treatment regimens may provide unique benefits.


Subject(s)
Antiviral Agents/pharmacology , Hepatitis C, Chronic/immunology , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Polyethylene Glycols/pharmacology , Ribavirin/pharmacology , Antigen Presentation , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Cells, Cultured , Dendritic Cells/immunology , Drug Therapy, Combination , Hepatitis C, Chronic/drug therapy , Humans , Interferon alpha-2 , Interferon-alpha/pharmacokinetics , Interferon-alpha/therapeutic use , Interferon-gamma/pharmacokinetics , Interferon-gamma/therapeutic use , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Plasma Cells/immunology , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/therapeutic use , Recombinant Proteins , Ribavirin/pharmacokinetics , Ribavirin/therapeutic use , Th1 Cells/immunology
18.
Curr Opin Microbiol ; 5(1): 87-91, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11834375

ABSTRACT

Chlamydia trachomatis is the most common cause of bacterial sexually transmitted disease in the United States, as well as the leading cause of preventable blindness worldwide. Immunity to C. trachomatis requires a variety of cell types, each employing an array of effector functions. Recent work has demonstrated that both CD4+ and CD8+ T lymphocytes play a major role in protective immunity to C. trachomatis, predominantly through their secretion of interferon-gamma. This review describes the generation of acquired immunity to C. trachomatis and focuses on how T cells contribute to both protection and immunopathology.


Subject(s)
Antigens, Bacterial/chemistry , Antigens, Bacterial/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Chlamydia trachomatis/growth & development , Chlamydia trachomatis/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interferon-gamma/pharmacokinetics , T-Lymphocytes/immunology , Adjuvants, Immunologic , Animals , Antibody Formation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chlamydia Infections/prevention & control , Humans , Immunity, Active , T-Lymphocytes/physiology
19.
Cancer Res ; 48(8): 2021-4, 1988 Apr 15.
Article in English | MEDLINE | ID: mdl-3127042

ABSTRACT

Recombinant murine gamma-interferon (rIFN-gamma) was radiolabeled by a novel procedure which does not require the use of preiodinated Bolton-Hunter reagent (specific activities of 0.5-3.0 microCi/micrograms). Gel filtration chromatography of the radiolabeled preparation yielded two peaks. The early eluting peak contained disulfide stabilized aggregates with minimal interferon antiviral activity. The second peak contained activity that was consistently greater than or equal to that of the nonradiolabeled rIFN-gamma. Two bands with apparent molecular weights of 17,000 and 34,000 were observed when the second peak was analyzed by SDS gel electrophoresis. Fractions comprising each of the two chromatography peaks were pooled separately and subjected to gel filtration again on identical columns 24 h after completion of the first column run. The elution volumes of each peak remained unchanged suggesting that the two forms are not in rapid equilibrium. The plasma clearance rates of [125I]rIFN-gamma before and after purification by chromatography were initially rapid but multiphasic. The slower phases of clearance did not result from stable association of the rIFN-gamma with plasma proteins. In organ distribution studies, the liver and spleen sequestered significant amounts of [125I]rIFN-gamma; however, the highest concentration of rIFN-gamma was recovered in the kidneys. A functional nephrectomy procedure was used to further study the role of the kidneys in rIFN-gamma clearance. Eliminating the kidneys significantly increased the amount of rIFN-gamma retained in the circulation, particularly at later times when the vascular [125I]rIFN-gamma levels were approximately threefold higher than in nonnephrectomized mice.


Subject(s)
Interferon-gamma/pharmacokinetics , Animals , Female , Kidney/metabolism , Metabolic Clearance Rate , Mice , Recombinant Proteins/pharmacokinetics , Tissue Distribution
20.
Cancer Res ; 51(24): 6643-9, 1991 Dec 15.
Article in English | MEDLINE | ID: mdl-1742738

ABSTRACT

We have investigated the action of recombinant human gamma-interferon (rHuIFN-gamma) against human ovarian cancer xenografts growing as ascites or as bulky solid i.p. tumors in nude mice. Both forms of the disease responded to i.p. rHuIFN-gamma with significant increases in mouse survival time, and in 2 of 3 ascitic models the mice were cured of peritoneal disease. The activity of rHuIFN-gamma was dose and schedule dependent, and xenografts derived from 3 different patients showed a heterogeneity of response. Peak i.p. levels of rHuIFN-gamma in nude mice bearing multiple i.p. solid tumors were similar to those found in ovarian cancer patients receiving i.p. rHuIFN-gamma, but clearance was more rapid in the mice. Rat gamma-interferon had no antitumor activity at the same doses and schedules although it had some biological activity in the nude mice. Histological examination of treated tumors revealed increased necrosis and loss of cellular organization with large areas of hypocellular epithelial mucin. These changes were preceded by a fall in tumor tryptophan and a rise in tumor kynurenine. We conclude that rHuIFN-gamma has a direct dose related antitumor effect on ovarian cancer xenografts that is preceded by increased metabolism of tryptophan.


Subject(s)
Cystadenocarcinoma/drug therapy , Interferon-gamma/therapeutic use , Ovarian Neoplasms/drug therapy , Animals , Ascites , Cystadenocarcinoma/metabolism , Cystadenocarcinoma/pathology , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Injections, Intraperitoneal , Interferon-gamma/administration & dosage , Interferon-gamma/pharmacokinetics , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Recombinant Proteins , Species Specificity , Survival Analysis , Transplantation, Heterologous , Tryptophan/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL