Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Blood ; 142(1): 62-72, 2023 07 06.
Article in English | MEDLINE | ID: mdl-36796019

ABSTRACT

Bruton tyrosine kinase (BTK), a nonreceptor tyrosine kinase, is a major therapeutic target for B-cell-driven malignancies. However, approved covalent BTK inhibitors (cBTKis) are associated with treatment limitations because of off-target side effects, suboptimal oral pharmacology, and development of resistance mutations (eg, C481) that prevent inhibitor binding. Here, we describe the preclinical profile of pirtobrutinib, a potent, highly selective, noncovalent (reversible) BTK inhibitor. Pirtobrutinib binds BTK with an extensive network of interactions to BTK and water molecules in the adenosine triphosphate binding region and shows no direct interaction with C481. Consequently, pirtobrutinib inhibits both BTK and BTK C481 substitution mutants in enzymatic and cell-based assays with similar potencies. In differential scanning fluorimetry studies, BTK bound to pirtobrutinib exhibited a higher melting temperature than cBTKi-bound BTK. Pirtobrutinib, but not cBTKis, prevented Y551 phosphorylation in the activation loop. These data suggest that pirtobrutinib uniquely stabilizes BTK in a closed, inactive conformation. Pirtobrutinib inhibits BTK signaling and cell proliferation in multiple B-cell lymphoma cell lines, and significantly inhibits tumor growth in human lymphoma xenografts in vivo. Enzymatic profiling showed that pirtobrutinib was highly selective for BTK in >98% of the human kinome, and in follow-up cellular studies pirtobrutinib retained >100-fold selectivity over other tested kinases. Collectively, these findings suggest that pirtobrutinib represents a novel BTK inhibitor with improved selectivity and unique pharmacologic, biophysical, and structural attributes with the potential to treat B-cell-driven cancers with improved precision and tolerability. Pirtobrutinib is being tested in phase 3 clinical studies for a variety of B-cell malignancies.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase , Lymphoma , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Humans , Animals , Xenograft Model Antitumor Assays , Lymphoma/drug therapy , Drug Evaluation, Preclinical , Cell Line, Tumor , Mice, Inbred NOD , Male , Mice, SCID , Molecular Conformation , Mice
2.
Protein J ; 39(2): 160-173, 2020 04.
Article in English | MEDLINE | ID: mdl-32172395

ABSTRACT

Previously we reported that site-specific modification of the human granulocyte-macrophage colony-stimulating factor (GM-CSF) A3C analog with polyethylene glycol (PEG) dramatically improved the pharmacokinetic properties of the protein in rats. However, we could not evaluate the hematological properties of the PEG-A3C protein in rats because human GM-CSF is inactive in rodents. To study the biological effects of PEGylated GM-CSF analogs in rodents we created a homologous site-specific PEGylated murine (mu) GM-CSF (T3C) protein. muGM-CSF and the T3C protein were expressed in Escherichia coli and purified by column chromatography. The purified T3C protein was covalently modified with a linear 20 kDa- or a branched 40 kDa-maleimide-PEG, and the monoPEGylated proteins purified by column chromatography. muGM-CSF, T3C and the two PEG-T3C proteins had comparable in vitro biological activities, as measured by stimulation of proliferation of the murine FDC-P1 cell line. The PEG-T3C proteins had 10- to 25-fold longer circulating half-lives than muGM-CSF and stimulated greater and longer lasting increases in neutrophils and white blood cells than muGM-CSF following a single intravenous or subcutaneous administration to rats. Treatment of rats made neutropenic with cyclophosphamide with the PEG-T3C proteins shortened the time for recovery of neutrophils to normal levels from 9 or 10 days to 5 or 6 days, whereas muGM-CSF showed no benefit versus vehicle solution. Acceleration of neutrophil recovery in cyclophosphamide-treated rats required a minimum of three PEG-T3C treatments over five days. The PEG-T3C proteins should prove useful for evaluating the potential therapeutic benefits of GM-CSF and long-acting GM-CSF proteins in rodent disease models.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacokinetics , Hematopoiesis/drug effects , Polyethylene Glycols/pharmacokinetics , Animals , Cell Line , Granulocyte Colony-Stimulating Factor/administration & dosage , Half-Life , Male , Neutropenia/drug therapy , Polyethylene Glycols/administration & dosage , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics
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.
J Interferon Cytokine Res ; 33(12): 769-77, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23962003

ABSTRACT

Interferon beta (IFN-ß) is widely used to ameliorate disease progression in patients with Multiple Sclerosis. IFN-ß has a short half-life in humans, necessitating frequent administration for optimum effectiveness. Covalent modification of IFN-ß with polyethylene glycol (PEG) improves the pharmacokinetic properties of the protein, but can adversely affect the protein's in vitro bioactivity. Random modification of lysine residues in IFN-ß with amine-reactive PEGs decreased the in vitro bioactivity of the protein 50-fold, presumably due to modification of lysine residues near critical receptor binding sites. PEGylated IFN-ß proteins that retained high in vitro bioactivity could be obtained by selective modification of the N-terminus of the protein with PEG. Here we use site-specific PEGylation technology (targeted attachment of a cysteine-reactive-PEG to an engineered cysteine residue in IFN-ß) to identify several additional amino acid positions where PEG can be attached to IFN-ß without appreciable loss of in vitro bioactivity. Unexpectedly, we found that most of the PEG-IFN-ß analogs showed 11- to 78-fold improved in vitro bioactivities relative to their unPEGylated parent proteins and to IFN-ß-1b. In vivo studies showed that a lead PEG-IFN-ß protein had improved pharmacokinetic properties compared to IFN-ß and was significantly more effective than IFN-ß at inhibiting growth of a human tumor xenograft in athymic mice.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Interferon-beta/chemistry , Interferon-beta/pharmacology , Polyethylene Glycols/chemistry , Amino Acid Substitution , Animals , Antineoplastic Agents/pharmacokinetics , Cell Proliferation/drug effects , Cysteine/chemistry , Dose-Response Relationship, Drug , Humans , Interferon beta-1b , Interferon-beta/pharmacokinetics , Male , Mice , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Xenograft Model Antitumor Assays
5.
Biotechnol Prog ; 26(3): 743-9, 2010.
Article in English | MEDLINE | ID: mdl-20196161

ABSTRACT

We expressed recombinant murine growth hormone (rmGH) in E. coli as a cost-effective way to produce large quantities (gram scale) of the protein for use in murine studies of immunogenicity to therapeutic proteins. High hydrostatic pressure was used to achieve high solubility and high refolding yields of rmGH protein produced in E. coli inclusion bodies. A two-step column purification protocol was used to produce 99% pure monomeric rmGH. Secondary and tertiary structures of purified rmGH were investigated using circular dichroism and 2D-UV spectroscopy. The purified rmGH produced was found to be biologically active in hypophysectomized rats.


Subject(s)
Escherichia coli/genetics , Growth Hormone/biosynthesis , Growth Hormone/chemistry , Inclusion Bodies/genetics , Animals , Chromatography, Gel , Chromatography, Ion Exchange , Circular Dichroism , Electrophoresis, Polyacrylamide Gel , Escherichia coli/chemistry , Escherichia coli/metabolism , Fermentation , Growth Hormone/genetics , Growth Hormone/metabolism , Hypophysectomy , Inclusion Bodies/chemistry , Inclusion Bodies/metabolism , Male , Mice , Protein Folding , Rats , Rats, Sprague-Dawley , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Solubility
6.
Curr Pharm Biotechnol ; 10(4): 447-55, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19519422

ABSTRACT

Non-denaturing pressures of around 2000 bar are effective for eliminating and refolding protein aggregates and may be applicable in various phases of protein manufacturing to decrease aggregate levels in products and improve process yields. Lower aggregate levels can result in reduced immunogenicity of proteins and enable the correct refolding of proteins that might not be recovered with traditional techniques. High pressure treatment can also be used to conduct selective PEGylation and protease cleavage reactions while minimizing protein aggregation. High pressure processes have been used in the food industry for over 50 years and large scale (300 L) systems are commercially available, enabling production of proteins on the kilogram scale. This review summarizes the utility of high pressure refolding to remove and refold protein aggregates, enhance therapeutic proteins, and facilitate manufacturing improvements at industrial scales.


Subject(s)
Biopharmaceutics/methods , Chemical Fractionation/methods , Multiprotein Complexes/chemistry , Multiprotein Complexes/isolation & purification , Proteins/chemistry , Proteins/isolation & purification , Technology, Pharmaceutical/methods , Pressure , Protein Denaturation , Protein Folding
7.
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
8.
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
9.
Exp Hematol ; 34(6): 697-704, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16728273

ABSTRACT

OBJECTIVE: Erythropoietin (Epo) bioactivity is significantly reduced by modification of lysine residues with amine-reactive reagents, which are the most commonly used reagents for attaching polyethylene glycols (PEGs) to proteins to improve protein half-life in vivo. The aims of this study were to determine whether Epo bioactivity can be preserved by targeting attachment of maleimide-PEGs to engineered cysteine analogs of Epo, and to determine whether the pegylated Epo cysteine analogs have improved pharmacokinetic properties in vivo. MATERIALS AND METHODS: Thirty-four Epo cysteine analogs were constructed by site-directed mutagenesis and expressed as secreted proteins in baculovirus-infected insect cells. Following purification, monopegylated derivatives of 12 cysteine analogs were prepared using 20-kDa maleimide-PEGs. In vitro biological activities of the proteins were measured in an Epo-dependent cell proliferation assay. Plasma levels of insect cell-expressed wild-type Epo (BV Epo) and a pegylated Epo cysteine analog were quantitated by ELISA following intravenous administration to rats. RESULTS: Biological activities of 17 purified Epo cysteine analogs and 10 purified pegylated Epo cysteine analogs were comparable to that of BV Epo in the in vitro bioassay. The only pegylated cysteine analogs that displayed consistently reduced in vitro bioactivities were substitutions for lysine residues, PEG-K45C and PEG-K154C. The pegylated Epo cysteine analog had a slower initial distribution phase and a longer terminal half-life than BV Epo in rats, but the majority of both proteins were cleared rapidly from the circulation. CONCLUSIONS: Targeted attachment of maleimide-PEGs to engineered Epo cysteine analogs permits rational design of monopegylated Epo analogs with minimal loss of in vitro biological activity. Insect cell-expressed Epo proteins are cleared rapidly from the circulation in rats, possibly due to improper glycosylation. Site-specific pegylation appears to improve the pharmacokinetic properties of Epo.


Subject(s)
Amino Acid Substitution , Cysteine/chemistry , Erythropoietin/chemistry , Ethylene Glycols/chemistry , Maleimides/chemistry , Animals , Biological Assay , Cell Line , Cysteine/genetics , Erythropoietin/administration & dosage , Erythropoietin/genetics , Erythropoietin/pharmacokinetics , Humans , Male , Rats , Rats, Sprague-Dawley , Recombinant Proteins
10.
Bioconjug Chem ; 16(5): 1291-8, 2005.
Article in English | MEDLINE | ID: mdl-16173810

ABSTRACT

Granulocyte macrophage colony-stimulating factor (GM-CSF) stimulates proliferation of hematopoietic cells of the macrophage and granulocyte lineages and is used clinically to treat neutropenia and other myeloid disorders. Because of its short circulating half-life, GM-CSF is administered to patients by daily injection. We describe here the engineering of highly potent, long-acting human GM-CSF proteins through site-specific modification of GM-CSF cysteine analogues with a cysteine-reactive poly(ethylene glycol) (PEG) reagent. Thirteen cysteine analogues of GM-CSF were constructed, primarily in nonhelical regions of the protein believed to lie away from the major receptor binding sites. The GM-CSF cysteine analogues were properly processed but insoluble following secretion into the Escherichia coli periplasm. The proteins were refolded and purified by column chromatography. Ten of the cysteine analogues could be modified with a 5-kDa maleimide PEG, and seven of the mono-PEGylated proteins were purified by ion-exchange column chromatography. Biological activities of the 13 cysteine analogues and 7 PEGylated cysteine analogues were comparable to that of wild-type GM-CSF in an in vitro cell proliferation assay using human TF-1 cells. One cysteine analogue was modified with larger 10-, 20-, and 40-kDa PEGs, with only minimal loss of in vitro bioactivity. Pharmacokinetic experiments in rats demonstrated that the PEGylated proteins had up to 47-fold longer circulating half-lives than wild-type GM-CSF. These data demonstrate the utility of site-specific PEGylation for creating highly potent, long-acting GM-CSF analogues and provide further evidence that the nonhelical regions of human GM-CSF examined are largely nonessential for biological activity of the protein.


Subject(s)
Cysteine/analogs & derivatives , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Polyethylene Glycols/chemistry , Animals , Cell Line , Cysteine/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/blood , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacokinetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Male , Protein Sorting Signals , Rats , Rats, Sprague-Dawley , Recombinant Proteins
11.
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
SELECTION OF CITATIONS
SEARCH DETAIL