Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
MAbs ; 15(1): 2256745, 2023.
Article in English | MEDLINE | ID: mdl-37698932

ABSTRACT

Biologic drug discovery pipelines are designed to deliver protein therapeutics that have exquisite functional potency and selectivity while also manifesting biophysical characteristics suitable for manufacturing, storage, and convenient administration to patients. The ability to use computational methods to predict biophysical properties from protein sequence, potentially in combination with high throughput assays, could decrease timelines and increase the success rates for therapeutic developability engineering by eliminating lengthy and expensive cycles of recombinant protein production and testing. To support development of high-quality predictive models for antibody developability, we designed a sequence-diverse panel of 83 effector functionless IgG1 antibodies displaying a range of biophysical properties, produced and formulated each protein under standard platform conditions, and collected a comprehensive package of analytical data, including in vitro assays and in vivo mouse pharmacokinetics. We used this robust training data set to build machine learning classifier models that can predict complex protein behavior from these data and features derived from predicted and/or experimental structures. Our models predict with 87% accuracy whether viscosity at 150 mg/mL is above or below a threshold of 15 centipoise (cP) and with 75% accuracy whether the area under the plasma drug concentration-time curve (AUC0-672 h) in normal mouse is above or below a threshold of 3.9 × 106 h x ng/mL.


Subject(s)
Antibodies, Monoclonal , Drug Discovery , Animals , Mice , Antibodies, Monoclonal/chemistry , Computer Simulation , Recombinant Proteins , Viscosity
2.
J Biol Chem ; 293(38): 14678-14688, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30068552

ABSTRACT

Fibroblast growth factor 21 (FGF21), an endocrine hormone in the FGF family, plays a critical role in regulating metabolic homeostasis and has emerged as a therapeutic target for metabolic diseases, including Type 2 diabetes mellitus. FGF21 functions through a receptor complex that consists of an FGF receptor (FGFR) and a co-receptor ß-Klotho. Here, we identify and biochemically and structurally characterize 39F7, a high-affinity agonistic monoclonal antibody (mAb) against ß-Klotho that mimics FGF21 function. The co-crystal structure of ß-Klotho KL1 domain in complex with 39F7 Fab revealed that the recognition of 39F7 is centered on Trp-295 of ß-Klotho in a FGF21 noncompetitive manner. KL1 adopts a (ß/α)8 TIM barrel fold which resembles that of ß-glycosylceramidase, but lacks molecular features for enzymatic activity, suggesting that KL1 functions as a scaffold protein instead. In vitro characterization demonstrated that, although 39F7 does not compete with FGF21, it is specific for ß-Klotho/FGFR1c activation. Furthermore, the agonistic activity of 39F7 required the full IgG molecule to be bivalent, suggesting that 39F7 functions by promoting receptor/co-receptor dimerization. Supported by negative stain EM analysis of full-length ß-Klotho, we propose a molecular model wherein the agonistic antibody 39F7 acts in a ß-Klotho- and FGFR1c-dependent manner, mimicking FGF21 activity. More importantly, 39F7 offers promising therapeutic potential in the axis of FGF21 signaling as an antibody therapy alternative to FGF21 analogs for treatment of metabolic diseases.


Subject(s)
Antibodies, Monoclonal/immunology , Fibroblast Growth Factors/metabolism , Membrane Proteins/immunology , Animals , Antibody Affinity , CHO Cells , Cricetulus , Crystallography, X-Ray , Humans , Klotho Proteins , Membrane Proteins/agonists , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Microscopy, Electron , Protein Binding , Protein Conformation , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction
3.
Sci Rep ; 8(1): 7570, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29765112

ABSTRACT

Small molecules and antibodies each have advantages and limitations as therapeutics. Here, we present for the first time to our knowledge, the structure-guided design of "chemibodies" as small molecule-antibody hybrids that offer dual recognition of a single target by both a small molecule and an antibody, using DPP-IV enzyme as a proof of concept study. Biochemical characterization demonstrates that the chemibodies present superior DPP-IV inhibition compared to either small molecule or antibody component alone. We validated our design by successfully solving a co-crystal structure of a chemibody in complex with DPP-IV, confirming specific binding of the small molecule portion at the interior catalytic site and the Fab portion at the protein surface. The discovery of chemibodies presents considerable potential for novel therapeutics that harness the power of both small molecule and antibody modalities to achieve superior specificity, potency, and pharmacokinetic properties.


Subject(s)
Dipeptidyl Peptidase 4/chemistry , Enzyme Inhibitors/chemical synthesis , Immunoconjugates/pharmacology , Animals , Antibodies, Monoclonal/chemistry , Catalytic Domain , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Immunoconjugates/chemistry , Models, Molecular , Rats , Small Molecule Libraries/chemistry , Structure-Activity Relationship
4.
Bioorg Med Chem Lett ; 25(23): 5546-50, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26526214

ABSTRACT

In this Letter, we report the continued optimization of the N-acyl-2-aminobenzimidazole series, focusing in particular on the N-alkyl substituent and 5-position of the benzimidazole based on the binding mode and the early SAR. These efforts led to the discovery of 16, a highly potent, selective, and orally bioavailable inhibitor of IRAK-4.


Subject(s)
Drug Discovery , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Administration, Oral , Animals , Benzimidazoles/chemistry , Enzyme Activation/drug effects , Molecular Structure , Protein Binding/drug effects , Protein Kinase Inhibitors/chemistry , Rats , Structure-Activity Relationship
5.
Bioorg Med Chem Lett ; 24(13): 2885-91, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24835984

ABSTRACT

Retinol-Binding Protein 4 (RBP4) is a plasma protein that transports retinol (vitamin A) from the liver to peripheral tissues. This Letter highlights our efforts in discovering the first, to our knowledge, non-retinoid small molecules that bind to RBP4 at the retinol site and reduce serum RBP4 levels in mice, by disrupting the interaction between RBP4 and transthyretin (TTR), a plasma protein that binds RBP4 and protects it from renal excretion. Potent compounds were discovered and optimized quickly from high-throughput screen (HTS) hits utilizing a structure-based approach. Inhibitor co-crystal X-ray structures revealed unique disruptions of RBP4-TTR interactions by our compounds through induced loop conformational changes instead of steric hindrance exemplified by fenretinide. When administered to mice, A1120, a representative compound in the series, showed concentration-dependent retinol and RBP4 lowering.


Subject(s)
Drug Discovery , Retinol-Binding Proteins, Plasma/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Ligands , Male , Mice , Models, Molecular , Molecular Structure , Rats , Retinol-Binding Proteins, Plasma/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Vitamin A/blood
6.
Structure ; 21(5): 798-809, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23602659

ABSTRACT

Sphingosine kinase 1 (SphK1) is a lipid kinase that catalyzes the conversion of sphingosine to sphingosine-1-phosphate (S1P), which has been shown to play a role in lymphocyte trafficking, angiogenesis, and response to apoptotic stimuli. As a central enzyme in modulating the S1P levels in cells, SphK1 emerges as an important regulator for diverse cellular functions and a potential target for drug discovery. Here, we present the crystal structures of human SphK1 in the apo form and in complexes with a substrate sphingosine-like lipid, ADP, and an inhibitor at 2.0-2.3 Å resolution. The SphK1 structures reveal a two-domain architecture in which its catalytic site is located in the cleft between the two domains and a hydrophobic lipid-binding pocket is buried in the C-terminal domain. Comparative analysis of these structures with mutagenesis and kinetic studies provides insight into how SphK1 recognizes the lipid substrate and catalyzes ATP-dependent phosphorylation.


Subject(s)
Lysophospholipids/chemistry , Sphingosine/analogs & derivatives , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Catalysis , Crystallography, X-Ray , Humans , Kinetics , Lysophospholipids/metabolism , Molecular Sequence Data , Phosphorylation , Protein Conformation , Sphingosine/chemistry , Sphingosine/metabolism , Substrate Specificity
7.
J Biol Chem ; 288(2): 1307-16, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23184939

ABSTRACT

Dipeptidyl peptidase IV (DPP-IV) degrades the incretin hormone glucagon-like peptide 1 (GLP-1). Small molecule DPP-IV inhibitors have been used as treatments for type 2 diabetes to improve glucose tolerance. However, each of the marketed small molecule drugs has its own limitation in terms of efficacy and side effects. To search for an alternative strategy of inhibiting DPP-IV activity, we generated a panel of tight binding inhibitory mouse monoclonal antibodies (mAbs) against rat DPP-IV. When tested in vitro, these mAbs partially inhibited the GLP-1 cleavage activity of purified enzyme and rat plasma. To understand the partial inhibition, we solved the co-crystal structure of one of the mAb Fabs (Ab1) in complex with rat DPP-IV. Although Ab1 does not bind at the active site, it partially blocks the side opening, which prevents the large substrates such as GLP-1 from accessing the active site, but not small molecules such as sitagliptin. When Ab1 was tested in vivo, it reduced plasma glucose and increased plasma GLP-1 concentration during an oral glucose tolerance test in rats. Together, we demonstrated the feasibility of using mAbs to inhibit DPP-IV activity and to improve glucose tolerance in a diabetic rat model.


Subject(s)
Antibodies, Monoclonal/immunology , Dipeptidyl Peptidase 4/immunology , Glucose Tolerance Test , Animals , Antibodies, Monoclonal/chemistry , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Mice , Mice, Inbred C57BL , Models, Molecular , Rats , Rats, Zucker
8.
J Biol Chem ; 287(33): 27326-34, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22718757

ABSTRACT

NF-κB-inducing kinase (NIK) is a central component in the non-canonical NF-κB signaling pathway. Excessive NIK activity is implicated in various disorders, such as autoimmune conditions and cancers. Here, we report the first crystal structure of truncated human NIK in complex with adenosine 5'-O-(thiotriphosphate) at a resolution of 2.5 Å. This truncated protein is a catalytically active construct, including an N-terminal extension of 60 residues prior to the kinase domain, the kinase domain, and 20 residues afterward. The structure reveals that the NIK kinase domain assumes an active conformation in the absence of any phosphorylation. Analysis of the structure uncovers a unique role for the N-terminal extension sequence, which stabilizes helix αC in the active orientation and keeps the kinase domain in the catalytically competent conformation. Our findings shed light on the long-standing debate over whether NIK is a constitutively active kinase. They also provide a molecular basis for the recent observation of gain-of-function activity for an N-terminal deletion mutant (ΔN324) of NIK, leading to constitutive non-canonical NF-κB signaling with enhanced B-cell adhesion and apoptosis resistance.


Subject(s)
Protein Serine-Threonine Kinases/chemistry , Thionucleotides/chemistry , Apoptosis/physiology , B-Lymphocytes/enzymology , Cell Adhesion/physiology , Cell Line , Crystallography, X-Ray , Humans , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Deletion , Thionucleotides/metabolism , NF-kappaB-Inducing Kinase
9.
J Med Chem ; 55(8): 3837-51, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22458568

ABSTRACT

The eukaryotic initiation factor 4E (eIF4E) plays a central role in the initiation of gene translation and subsequent protein synthesis by binding the 5' terminal mRNA cap structure. We designed and synthesized a series of novel compounds that display potent binding affinity against eIF4E despite their lack of a ribose moiety, phosphate, and positive charge as present in m7-GMP. The biochemical activity of compound 33 is 95 nM for eIF4E in an SPA binding assay. More importantly, the compound has an IC(50) of 2.5 µM for inhibiting cap-dependent mRNA translation in a rabbit reticular cell extract assay (RRL-IVT). This series of potent, truncated analogues could serve as a promising new starting point toward the design of neutral eIF4E inhibitors with physicochemical properties suitable for cellular activity assessment.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , Guanine/analogs & derivatives , Guanosine Monophosphate/analogs & derivatives , Guanosine Monophosphate/pharmacology , Organophosphonates/chemical synthesis , RNA Caps/metabolism , Animals , Crystallography, X-Ray , Drug Design , Eukaryotic Initiation Factor-4E/chemistry , Guanine/chemical synthesis , Guanine/pharmacology , Guanosine Monophosphate/chemical synthesis , Humans , Inhibitory Concentration 50 , Models, Molecular , Organophosphonates/pharmacology , Phosphorous Acids , Protein Biosynthesis/drug effects , RNA Caps/chemistry , Rabbits , Reticulocytes/drug effects , Reticulocytes/metabolism , Structure-Activity Relationship
11.
J Biol Chem ; 284(12): 7673-80, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19147488

ABSTRACT

Retinol-binding protein 4 (RBP4) transports retinol from the liver to extrahepatic tissues, and RBP4 lowering is reported to improve insulin sensitivity in mice. We have identified A1120, a high affinity (K(i) = 8.3 nm) non-retinoid ligand for RBP4, which disrupts the interaction between RBP4 and its binding partner transthyretin. Analysis of the RBP4-A1120 co-crystal structure reveals that A1120 induces critical conformational changes at the RBP4-transthyretin interface. Administration of A1120 to mice lowers serum RBP4 and retinol levels but, unexpectedly, does not improve insulin sensitivity. In addition, we show that Rpb4(-/-) mice display normal insulin sensitivity and are not protected from high fat diet-induced insulin resistance. We conclude that lowering RBP4 levels does not improve insulin sensitivity in mice. Therefore, RBP4 lowering may not be an effective strategy for treating diabetes.


Subject(s)
Heterocyclic Compounds, 3-Ring/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Piperidines/chemistry , Retinol-Binding Proteins, Plasma , Vitamin A/blood , Animals , Crystallography, X-Ray , Diabetes Mellitus/blood , Diabetes Mellitus/drug therapy , Dietary Fats/adverse effects , Humans , Insulin/metabolism , Insulin Resistance , Ligands , Mice , Mice, Knockout , Piperidines/pharmacology , Protein Structure, Tertiary , Retinol-Binding Proteins, Plasma/agonists , Retinol-Binding Proteins, Plasma/chemistry , Retinol-Binding Proteins, Plasma/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...