Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Radiat Res ; 202(1): 26-37, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38714310

ABSTRACT

BBT-059, a long-acting PEGylated interleukin-11 (IL-11) analog that is believed to have hematopoietic promoting and anti-apoptotic properties, is being developed as a potential radiation medical countermeasure (MCM) for hematopoietic acute radiation syndrome (H-ARS). This agent has been shown to improve survival in lethally irradiated mice. To further evaluate the drug's toxicity and safety profile, 12 naïve nonhuman primates (NHPs, rhesus macaques) were administered one of three doses of BBT-059 subcutaneously and were monitored for the next 21 days. Blood samples were collected throughout the study to assess the pharmacokinetics (PK) and pharmacodynamics (PD) of the drug as well as its effects on complete blood counts, cytokines, vital signs, and to conduct metabolomic studies. No adverse effects were detected in any treatment group during the study. Short-term changes in metabolomic profiles were present in all groups treated with BBT-059 beginning immediately after drug administration and reverting to near normal levels by the end of the study period. Several pathways and metabolites, particularly those related to inflammation and steroid hormone biosynthesis, were activated by BBT-059 administration. Taken together, these observations suggest that BBT-059 has a good safety profile for further development as a radiation MCM for regulatory approval for human use.


Subject(s)
Macaca mulatta , Metabolomics , Polyethylene Glycols , Radiation-Protective Agents , Animals , Radiation-Protective Agents/pharmacology , Radiation-Protective Agents/pharmacokinetics , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/pharmacology , Polyethylene Glycols/chemistry , Male , Interleukin-11 , Female , Metabolome/drug effects , Metabolome/radiation effects , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/prevention & control
2.
Protein Eng Des Sel ; 28(10): 461-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25855658

ABSTRACT

Crohn's Disease (CD) afflicts over half a million Americans with an annual economic impact exceeding $10 billion. Granulocyte macrophage colony-stimulating factor (GM-CSF) can increase patient immune responses against intestinal microbes that promote CD and has been effective for some patients in clinical trials. We have made important progress toward developing GM-CSF variants that could be more effective CD therapeutics by virtue of being less prone to neutralization by the endogenous GM-CSF autoantibodies that are highly expressed in CD patients. Yeast display engineering revealed mutations that increase GM-CSF variant binding affinity by up to ∼3-fold toward both GM-CSF receptor alpha and beta subunits in surface plasmon resonance experiments. Increased binding affinity did not reduce GM-CSF half-maximum effective concentration (EC50) values in conventional in vitro human leukocyte proliferation assays. Affinity-enhancing mutations did, however, promote a 'refacing effect' that imparted all five evaluated GM-CSF variants with increased in vitro bioactivity in the presence of GM-CSF-neutralizing polyclonal antisera. The most improved variant, H15L/R23L, was 6-fold more active than wild-type GM-CSF. Incorporation of additional known affinity-increasing mutations could augment the refacing effect and concomitant bioactivity improvements described here.


Subject(s)
Antibodies, Neutralizing/immunology , Cytokines/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Protein Engineering , Cell Proliferation , Cytokine Receptor Common beta Subunit/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Humans , Leukocytes/cytology , Models, Molecular , Mutation , Protein Structure, Secondary , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology
3.
J Interferon Cytokine Res ; 34(10): 759-68, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24841172

ABSTRACT

Interferon gamma (IFN-γ) is a 28 kDa homodimeric cytokine that exhibits potent immunomodulatory, anti-proliferative, and antiviral properties. The protein is used to treat chronic granulomatous disease and malignant osteopetrosis, and it is under investigation as a treatment for a variety of cancer, fungal and viral diseases. IFN-γ has a short circulating half life in vivo, which necessitates frequent administration to patients. An unusual feature of IFN-γ is that the protein contains no native cysteines. To create a longer-acting and potentially more effective form of the protein, we introduced a cysteine residue into the IFN-γ coding sequence at amino acid position 103, which is located in a surface-exposed, non-helical region of the protein. The added cysteine residue served as the site for targeted modification of the protein with a cysteine-reactive polyethylene glycol (PEG) reagent. The recombinant protein was expressed in bacteria, purified and modified with 10, 20, and 40 kDa maleimide PEGs. The purified, PEGylated proteins had in vitro bioactivities comparable to IFN-γ, as measured using an in vitro cell growth inhibition assay. The PEGylated proteins displayed 20- to 32-fold longer half lives than IFN-γ in rats, and they were significantly more effective than IFN-γ at inhibiting growth of a human tumor xenograft in athymic mice.


Subject(s)
Cysteine/chemistry , Growth Inhibitors/pharmacokinetics , Interferon-gamma/pharmacokinetics , Polyethylene Glycols/chemistry , Animals , Cell Line, Tumor , Cysteine/genetics , Female , Growth Inhibitors/chemistry , Growth Inhibitors/pharmacology , Humans , Immunomodulation , Interferon-gamma/chemistry , Interferon-gamma/pharmacology , Male , Mice , Mice, Nude , Mutation/genetics , Protein Binding , Protein Conformation , Protein Engineering , Protein Stability , Rats , Rats, Sprague-Dawley , Xenograft Model Antitumor Assays
4.
PLoS One ; 9(3): e91990, 2014.
Article in English | MEDLINE | ID: mdl-24637521

ABSTRACT

Previously we showed that granulocyte colony-stimulating factor (G-CSF) in vitro bioactivity is preserved when the protein is joined via a flexible 7 amino acid linker to an immunoglobulin-1 (IgG1)-Fc domain and that the G-CSF/IgG1-Fc fusion protein possessed a longer circulating half-life and improved hematopoietic properties compared to G-CSF in normal rats. We have extended this analysis by comparing the relative hematopoietic potencies of G-CSF/IgG1-Fc to G-CSF in normal mice and to G-CSF and polyethylene glycol (PEG) -modified G-CSF in neutropenic rats. Mice were treated for 5 days using different doses and dosing regimens of G-CSF/IgG1-Fc or G-CSF and circulating neutrophil levels in the animals measured on Day 6. G-CSF/IgG1-Fc stimulated greater increases in blood neutrophils than comparable doses of G-CSF when administered using daily, every other day or every third day dosing regimens. In rats made neutropenic with cyclophosphamide, G-CSF/IgG1-Fc accelerated recovery of blood neutrophils to normal levels (from Day 9 to Day 5) when administered as 5 daily injections or as a single injection on Day 1. By contrast, G-CSF accelerated neutrophil recovery when administered as 5 daily injections, but not when administered as a single injection. G-CSF/IgG1-Fc was as effective as PEG-G-CSF at accelerating neutrophil recovery following a single injection in neutropenic rats. G-CSF/IgG1-Fc and G-CSF/IgG4-Fc fusion proteins in which the 7 amino acid linker was deleted also were effective at accelerating neutrophil recovery following a single injection in neutropenic rats. These studies confirm the enhanced in vivo hematopoietic properties of G-CSF/IgG-Fc fusion proteins.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoiesis/drug effects , Immunoglobulin Fc Fragments/pharmacology , Neutropenia/drug therapy , Recombinant Fusion Proteins/pharmacology , Animals , Blood Cell Count , Disease Models, Animal , Female , Gene Expression , Gene Order , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocyte Colony-Stimulating Factor/genetics , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/genetics , Leukocyte Count , Mice , Neutropenia/etiology , Neutrophils/drug effects , Rats , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Time Factors
5.
Bioconjug Chem ; 19(1): 299-305, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18020402

ABSTRACT

Recombinant interferon alpha-2 (IFN-alpha2) has proven useful for treating a variety of human cancers and viral diseases. IFN-alpha2 has a short circulating half-life in vivo, which necessitates daily or thrice weekly administration to patients. It is possible to extend the circulating half-life of IFN-alpha2 by random modification of lysine residues in the protein with polyethylene glycol (PEG); however, such preparations have heterogeneous structures and low specific activities, and may not provide optimal therapeutic benefits to patients. A long-acting, site-specific, monoPEGylated IFN-alpha2 protein has now been created by targeted attachment of a 20 kDa or a 40 kDa maleimide-PEG to a cysteine analogue of IFN-alpha2, M111C. In vitro bioactivities of the purified 20 kDa and 40 kDa PEG-M111C proteins were within 2- to 3-fold of those of wild type IFN-alpha2 and 7- to 10-fold better than that of a 40 kDa PEG IFN-alpha2 protein created using nontargeted, amine-PEGylation methodology. The 20 kDa and 40 kDa PEG-M111C proteins demonstrated 26- to 38-fold longer half-lives, respectively, than IFN-alpha2 following subcutaneous administration to rats. The 20 kDa PEG M111C protein inhibited growth of human NIH:OVCAR-3 cells transplanted into nude mice by >90%, as measured by tumor size, tumor weight, and number of animals with detectable tumors at necropsy, and was significantly more effective than a comparable dose of IFN-alpha2. These data extend our previous findings that bioactivity of IFN-alpha2 can be largely preserved by targeted attachment of PEG moieties to nonessential sites in the protein and provide evidence that site-specific PEGylated IFN-alpha2 proteins possess enhanced tumoricidal properties in vivo.


Subject(s)
Antineoplastic Agents/blood , Antineoplastic Agents/pharmacology , Interferon-alpha/blood , Interferon-alpha/pharmacology , Polyethylene Glycols/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Cysteine , Half-Life , Humans , Interferon-alpha/chemistry , Interferon-alpha/pharmacokinetics , Mice , Mice, Nude , Recombinant Proteins/blood , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Sensitivity and Specificity , Transplantation, Heterologous
6.
Endocrinology ; 148(4): 1590-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17234711

ABSTRACT

Recombinant human GH is used to treat GH deficiency in children and adults and wasting in AIDS patients. GH has a circulating half-life of only a few hours in humans and must be administered to patients by daily injection for maximum effectiveness. Previous studies showed that longer-acting forms of GH could be created by modification of GH with multiple 5-kDa amine-reactive polyethylene glycols (PEGs). Eight of nine lysine residues and the N-terminal amino acid were modified to varying extents by amine PEGylation of GH. The amine-PEGylated GH product comprised a complex mixture of multiple PEGylated species that differed from one another in mass, in vitro bioactivity, and in vivo potency. In vitro bioactivity of GH was reduced 100- to 1000-fold by extensive amine PEGylation of the protein. Here we describe a homogeneously modified, mono-PEGylated GH protein that possesses near complete in vitro bioactivity, a long half-life, and increased potency in vivo. The mono-PEGylated GH was created by substituting cysteine for threonine-3 (T3C) of GH, followed by modification of the added cysteine residue with a single 20-kDa cysteine-reactive PEG. The PEG-T3C protein has an approximate 8-fold longer half-life than GH after sc administration to rats. Every other day or every third day administration of PEG-T3C stimulates increases in body weight and tibial epiphysis growth comparable with that produced by daily administration of GH in hypophysectomized rats. Long-acting, mono-PEGylated GH analogs such as PEG-T3C are promising candidates for future testing in humans.


Subject(s)
Bone Development/drug effects , Human Growth Hormone/analogs & derivatives , Human Growth Hormone/pharmacokinetics , Hypophysectomy , Weight Gain/drug effects , Animals , Delayed-Action Preparations , Drug Evaluation, Preclinical , Half-Life , Human Growth Hormone/chemistry , Male , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Rats , Rats, Sprague-Dawley , Tibia/drug effects , Treatment Outcome
7.
Bioconjug Chem ; 16(1): 200-7, 2005.
Article in English | MEDLINE | ID: mdl-15656592

ABSTRACT

Recombinant interferon alpha-2 (IFN-alpha2) is used clinically to treat a variety of viral diseases and cancers. IFN-alpha2 has a short circulating half-life, which necessitates frequent administration to patients. Previous studies showed that it is possible to extend the circulating half-life of IFN-alpha2 by modifying lysine residues of the protein with amine-reactive poly(ethylene glycol) (PEG) reagents. However, amine-PEGylated IFN-alpha2 comprises a heterogeneous product mixture with low specific activity due to the large number and critical locations of lysine residues in IFN-alpha2. In an effort to overcome these problems we determined the feasibility of creating site-specific, mono-PEGylated IFN-alpha2 analogues by introducing a free (unpaired) cysteine residue into the protein, followed by modification of the added cysteine residue with a maleimide-PEG reagent. IFN-alpha2 cysteine analogues were expressed in Escherichia coli and purified, and their in vitro bioactivities were measured in the human Daudi cell line growth inhibition assay. Several cysteine analogues were identified that do not significantly affect in vitro biological activity of IFN-alpha2. Certain of the cysteine analogues, but not wild-type IFN-alpha2, reacted with maleimide-PEG to produce mono-PEGylated proteins. The PEG-Q5C analogue retained high in vitro bioactivity (within 3- to 4-fold of wild-type IFN-alpha2) even when modified with 20- and 40-kDa PEGs. Pharmacokinetic experiments indicated that the 20-kDa PEG-Q5C and 40-kDa PEG-Q5C proteins have 20-fold and 40-fold longer half-lives, respectively, than IFN-alpha2 following subcutaneous administration to rats. These studies demonstrate the feasibility of using site-specific PEGylation technology to create a long-acting, mono-PEGylated IFN-alpha2 protein with high specific activity.


Subject(s)
Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , Interferon-alpha/pharmacology , Polyethylene Glycols/chemistry , Animals , Base Sequence , Binding Sites , Cells, Cultured , Cysteine/chemistry , Dose-Response Relationship, Drug , Escherichia coli/genetics , Humans , Interferon-alpha/chemistry , Lysine/chemistry , Maleimides/chemistry , Maleimides/pharmacology , Molecular Weight , Proteins/chemistry , Rats
8.
Exp Hematol ; 32(5): 441-9, 2004 May.
Article in English | MEDLINE | ID: mdl-15145212

ABSTRACT

OBJECTIVE: The aim of this study was to determine whether fusion proteins comprising human granulocyte colony-stimulating factor (G-CSF) joined to human immunoglobulin G1 and G4 (IgG1 and IgG4) Fc and C(H) domains are biologically active and have improved pharmacokinetic and hematopoietic properties in vivo. MATERIAL AND METHODS: Chimeric genes encoding human G-CSF fused to the N-termini of the Fc and C(H) domains of human IgG1 and IgG4 were constructed and used to transfect monkey COS cells. The fusion proteins were purified from the conditioned media by protein A affinity chromatography. Bioactivities of the proteins were measured in a G-CSF-dependent in vitro bioassay. Pharmacokinetic and granulopoietic properties of the G-CSF/IgG1-Fc fusion protein were measured in normal rats. RESULTS: The G-CSF/IgG-Fc and G-CSF/IgG-C(H) fusion proteins were secreted from transfected COS cells primarily as disulfide-linked homodimers. On a molar basis, the purified G-CSF/IgG-Fc fusion proteins were as active as G-CSF in in vitro bioassays, whereas bioactivities of the purified G-CSF/IgG-C(H) fusion proteins were decreased 3- to 4-fold. The G-CSF/IgG1-Fc fusion protein displayed a slower plasma clearance rate and stimulated greater and longer lasting increases in circulating neutrophils and white blood cells than G-CSF following intravenous and subcutaneous administration to rats. CONCLUSION: Fusion of G-CSF to human IgG domains results in homodimeric fusion proteins possessing high in vitro bioactivities, long circulating half-lives, and enhanced hematopoietic properties in vivo.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoiesis/drug effects , Immunoglobulin G/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Blood Cell Count , Bone Marrow Examination , COS Cells , Granulocyte Colony-Stimulating Factor/blood , Granulocyte Colony-Stimulating Factor/genetics , Half-Life , Humans , Immunoglobulin G/genetics , Leukocytes/cytology , Leukocytes/drug effects , Male , Neutrophils/cytology , Neutrophils/drug effects , Pharmacokinetics , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...