Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Med Chem Lett ; 13(11): 1776-1782, 2022 Nov 10.
Article in English | MEDLINE | ID: mdl-36385934

ABSTRACT

The diastereomeric macrocyclic calcitonin gene-related peptide (CGRP) antagonists HTL0029881 (3) and HTL0029882 (4), in which the stereochemistry of a spiro center is reversed, surprisingly demonstrate comparable potency. X-ray crystallographic characterization demonstrates that 3 binds to the CGRP receptor in a precedented manner but that 4 binds in an unprecedented, unexpected, and radically different manner. The observation of this phenomenon is noteworthy and may open novel avenues for CGRP receptor antagonist design.

2.
ACS Chem Neurosci ; 13(6): 751-765, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35245037

ABSTRACT

A series of macrocyclic calcitonin gene-related peptide (CGRP) receptor antagonists identified using structure-based design principles, exemplified by HTL0028016 (1) and HTL0028125 (2), is described. Structural characterization by X-ray crystallography of the interaction of two of the macrocycle antagonists with the CGRP receptor ectodomain is described, along with structure-activity relationships associated with point changes to the macrocyclic antagonists. The identification of non-peptidic/natural product-derived, macrocyclic ligands for a G protein coupled receptor (GPCR) is noteworthy.


Subject(s)
Receptors, Calcitonin Gene-Related Peptide , Receptors, G-Protein-Coupled , Calcitonin Receptor-Like Protein/chemistry , Calcitonin Receptor-Like Protein/metabolism , Crystallography, X-Ray , Ligands , Receptors, Calcitonin Gene-Related Peptide/chemistry , Receptors, Calcitonin Gene-Related Peptide/metabolism , Receptors, G-Protein-Coupled/metabolism
3.
J Med Chem ; 63(14): 7906-7920, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32558564

ABSTRACT

Structure-based drug design enabled the discovery of 8, HTL22562, a calcitonin gene-related peptide (CGRP) receptor antagonist. The structure of 8 complexed with the CGRP receptor was determined at a 1.6 Å resolution. Compound 8 is a highly potent, selective, metabolically stable, and soluble compound suitable for a range of administration routes that have the potential to provide rapid systemic exposures with resultant high levels of receptor coverage (e.g., subcutaneous). The low lipophilicity coupled with a low anticipated clinically efficacious plasma exposure for migraine also suggests a reduced potential for hepatotoxicity. These properties have led to 8 being selected as a clinical candidate for acute treatment of migraine.


Subject(s)
Calcitonin Gene-Related Peptide Receptor Antagonists/pharmacology , Indazoles/pharmacology , Receptors, Calcitonin Gene-Related Peptide/metabolism , Spiro Compounds/pharmacology , Animals , Binding Sites , Calcitonin Gene-Related Peptide Receptor Antagonists/chemical synthesis , Calcitonin Gene-Related Peptide Receptor Antagonists/metabolism , Calcitonin Gene-Related Peptide Receptor Antagonists/toxicity , Dogs , Drug Design , Humans , Indazoles/chemical synthesis , Indazoles/metabolism , Indazoles/toxicity , Macaca fascicularis , Migraine Disorders/drug therapy , Molecular Docking Simulation , Molecular Structure , Rats , Spiro Compounds/chemical synthesis , Spiro Compounds/metabolism , Spiro Compounds/toxicity , Structure-Activity Relationship
4.
J Med Chem ; 63(4): 1528-1543, 2020 02 27.
Article in English | MEDLINE | ID: mdl-31860301

ABSTRACT

The orexin system, which consists of the two G protein-coupled receptors OX1 and OX2, activated by the neuropeptides OX-A and OX-B, is firmly established as a key regulator of behavioral arousal, sleep, and wakefulness and has been an area of intense research effort over the past two decades. X-ray structures of the receptors in complex with 10 new antagonist ligands from diverse chemotypes are presented, which complement the existing structural information for the system and highlight the critical importance of lipophilic hotspots and water molecules for these peptidergic GPCR targets. Learnings from the structural information regarding the utility of pharmacophore models and how selectivity between OX1 and OX2 can be achieved are discussed.


Subject(s)
Orexin Receptor Antagonists/metabolism , Orexin Receptors/metabolism , Binding Sites , Computer Simulation , Crystallography, X-Ray , HEK293 Cells , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Ligands , Orexin Receptor Antagonists/chemistry , Orexin Receptors/chemistry
5.
Biochem Pharmacol ; 147: 38-54, 2018 01.
Article in English | MEDLINE | ID: mdl-29102678

ABSTRACT

Recent interest has focused on antibodies that can discriminate between different receptor conformations. Here we have characterised the effect of a monoclonal antibody (mAb3), raised against a purified thermo-stabilised turkey ß1-adrenoceptor (ß1AR-m23 StaR), on ß1-ARs expressed in CHO-K1 or HEK 293 cells. Immunohistochemical and radioligand-binding studies demonstrated that mAb3 was able to bind to ECL2 of the tß1-AR, but not its human homologue. Specific binding of mAb3 to tß1-AR was inhibited by a peptide based on the turkey, but not the human, ECL2 sequence. Studies with [3H]-CGP 12177 demonstrated that mAb3 prevented the binding of orthosteric ligands to a subset (circa 40%) of turkey ß1-receptors expressed in both CHO K1 and HEK 293 cells. MAb3 significantly reduced the maximum specific binding capacity of [3H]-CGP-12177 without influencing its binding affinity. Substitution of ECL2 of tß1-AR with its human equivalent, or mutation of residues D186S, P187D, Q188E prevented the inhibition of [3H]-CGP 12177 binding by mAb3. MAb3 also elicited a negative allosteric effect on agonist-stimulated cAMP responses. The identity of the subset of turkey ß1-adrenoceptors influenced by mAb3 remains to be established but mAb3 should become an important tool to investigate the nature of ß1-AR conformational states and oligomeric complexes.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/metabolism , Adrenergic beta-Agonists/metabolism , Allosteric Regulation/physiology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , Propanolamines/metabolism , Protein Binding/physiology , Protein Stability , Protein Structure, Secondary , Receptors, Adrenergic, beta-1/genetics , Turkey
6.
MAbs ; 6(1): 246-61, 2014.
Article in English | MEDLINE | ID: mdl-24253107

ABSTRACT

Thermostabilized G protein-coupled receptors used as antigens for in vivo immunization have resulted in the generation of functional agonistic anti-ß1-adrenergic (ß1AR) receptor monoclonal antibodies (mAbs). The focus of this study was to examine the pharmacology of these antibodies to evaluate their mechanistic activity at ß1AR. Immunization with the ß1AR stabilized receptor yielded five stable hybridoma clones, four of which expressed functional IgG, as determined in cell-based assays used to evaluate cAMP stimulation. The antibodies bind diverse epitopes associated with low nanomolar agonist activity at ß1AR, and they appeared to show some degree of biased signaling as they were inactive in an assay measuring signaling through ß-arrestin. In vitro characterization also verified different antibody receptor interactions reflecting the different epitopes on the extracellular surface of ß1AR to which the mAbs bind. The anti-ß1AR mAbs only demonstrated agonist activity when in dimeric antibody format, but not as the monomeric Fab format, suggesting that agonist activation may be mediated through promoting receptor dimerization. Finally, we have also shown that at least one of these antibodies exhibits in vivo functional activity at a therapeutically-relevant dose producing an increase in heart rate consistent with ß1AR agonism.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Arrestins/immunology , Avian Proteins/immunology , Receptors, Adrenergic, beta-1/immunology , Signal Transduction/drug effects , Animals , Avian Proteins/agonists , Female , HEK293 Cells , Humans , Mice, Inbred BALB C , Signal Transduction/immunology , Turkeys , beta-Arrestins
7.
Proc Natl Acad Sci U S A ; 106(5): 1566-71, 2009 Feb 03.
Article in English | MEDLINE | ID: mdl-19164533

ABSTRACT

Human metapneumovirus (hMPV) is a recently described paramyxovirus that causes lower respiratory infections in children and adults worldwide. The hMPV fusion (F) protein is a membrane-anchored glycoprotein and major protective antigen. All hMPV F protein sequences determined to date contain an Arg-Gly-Asp (RGD) sequence, suggesting that F engages RGD-binding integrins to mediate cell entry. The divalent cation chelator EDTA, which disrupts heterodimeric integrin interactions, inhibits infectivity of hMPV but not the closely related respiratory syncytial virus (RSV), which lacks an RGD motif. Function-blocking antibodies specific for alphavbeta1 integrin inhibit infectivity of hMPV but not RSV. Transfection of nonpermissive cells with alphav or beta1 cDNAs confers hMPV infectivity, whereas reduction of alphav and beta1 integrin expression by siRNA inhibits hMPV infection. Recombinant hMPV F protein binds to cells, whereas Arg-Gly-Glu (RGE)-mutant F protein does not. These data suggest that alphavbeta1 integrin is a functional receptor for hMPV.


Subject(s)
Metapneumovirus/pathogenicity , Receptors, Vitronectin/physiology , Virulence/physiology , Animals , Antibodies, Viral/immunology , Humans , Metapneumovirus/immunology , RNA, Small Interfering , Receptors, Vitronectin/immunology , Swine , Transfection , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/physiology
8.
J Virol ; 81(15): 8315-24, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17522220

ABSTRACT

Human metapneumovirus (hMPV) is a recently discovered paramyxovirus that is a major cause of lower-respiratory-tract disease. hMPV is associated with more severe disease in infants and persons with underlying medical conditions. Animal studies have shown that the hMPV fusion (F) protein alone is capable of inducing protective immunity. Here, we report the use of phage display technology to generate a fully human monoclonal antibody fragment (Fab) with biological activity against hMPV. Phage antibody libraries prepared from human donor tissues were selected against recombinant hMPV F protein with multiple rounds of panning. Recombinant Fabs then were expressed in bacteria, and supernatants were screened by enzyme-linked immunosorbent assay and immunofluorescent assays. A number of Fabs that bound to hMPV F were isolated, and several of these exhibited neutralizing activity in vitro. Fab DS7 neutralized the parent strain of hMPV with a 60% plaque reduction activity of 1.1 mug/ml and bound to hMPV F with an affinity of 9.8 x10(-10) M, as measured by surface plasmon resonance. To test the in vivo activity of Fab DS7, groups of cotton rats were infected with hMPV and given Fab intranasally 3 days after infection. Nasal turbinates and lungs were harvested on day 4 postinfection and virus titers determined. Animals treated with Fab DS7 exhibited a >1,500-fold reduction in viral titer in the lungs, with a modest 4-fold reduction in the nasal tissues. There was a dose-response relationship between the dose of DS7 and virus titer. Human Fab DS7 may have prophylactic or therapeutic potential against severe hMPV infection.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulin Fab Fragments/immunology , Metapneumovirus/immunology , Paramyxoviridae Infections/therapy , Recombinant Proteins/immunology , Viral Fusion Proteins/immunology , Animals , Antibodies, Monoclonal/genetics , Cell Line , Epitopes , Humans , Immunoglobulin Fab Fragments/genetics , Infant , Metapneumovirus/genetics , Neutralization Tests , Peptide Library , Rats , Recombinant Proteins/genetics , Sigmodontinae , Viral Fusion Proteins/genetics , Viral Load
9.
J Virol ; 81(2): 698-707, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17050599

ABSTRACT

Human metapneumovirus (hMPV) is a recently described paramyxovirus that is a major cause of upper and lower respiratory infection in children and adults worldwide. A safe and effective vaccine could decrease the burden of disease associated with this novel pathogen. We previously reported the development of the cotton rat model of hMPV infection and pathogenesis (J. V. Williams et al., J. Virol. 79:10944-10951, 2005). We report here the immunogenicity of an hMPV fusion (F) protein in this model. We constructed DNA plasmids that exhibited high levels of expression of hMPV F in mammalian cells (DNA-F). These constructs were used to develop a novel strategy to produce highly pure, soluble hMPV F protein lacking the transmembrane domain (FDeltaTM). We then immunized cotton rats at 0 and 14 days with either control vector, DNA-F alone, DNA-F followed by FDeltaTM protein, or FDeltaTM alone. All groups were challenged intranasally at 28 days with live hMPV. All three groups that received some form of hMPV F immunization mounted neutralizing antibody responses and exhibited partial protection against virus shedding in the lungs compared to controls. The FDeltaTM-immunized animals showed the greatest degree of protection (>1,500-fold reduction in lung virus titer). All three immunized groups showed a modest reduction of nasal virus shedding. Neither evidence of a Th2-type response nor increased lung pathology were present in the immunized animals. We conclude that sequence-optimized hMPV F protein protects against hMPV infection when delivered as either a DNA or a protein vaccine in cotton rats.


Subject(s)
Metapneumovirus/immunology , Paramyxoviridae Infections/prevention & control , Respiratory Tract Infections/prevention & control , Viral Fusion Proteins , Viral Vaccines , Animals , Antibodies, Viral/blood , Cell Line , Disease Models, Animal , Humans , Immunization , Lung/pathology , Lung/virology , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/virology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Sigmodontinae , Viral Fusion Proteins/administration & dosage , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology , Viral Fusion Proteins/metabolism , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...