Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 15(681): eabq5241, 2023 02.
Article in English | MEDLINE | ID: mdl-36724238

ABSTRACT

In October 2019, Novartis launched brolucizumab, a single-chain variable fragment molecule targeting vascular endothelial growth factor A, for the treatment of neovascular age-related macular degeneration. In 2020, rare cases of retinal vasculitis and/or retinal vascular occlusion (RV/RO) were reported, often during the first few months after treatment initiation, consistent with a possible immunologic pathobiology. This finding was inconsistent with preclinical studies in cynomolgus monkeys that demonstrated no drug-related intraocular inflammation, or RV/RO, despite the presence of preexisting and treatment-emergent antidrug antibodies (ADAs) in some animals. In this study, the immune response against brolucizumab in humans was assessed using samples from clinical trials and clinical practice. In the brolucizumab-naïve population, anti-brolucizumab ADA responses were detected before any treatment, which was supported by the finding that healthy donors can harbor brolucizumab-specific B cells. This suggested prior exposure of the immune system to proteins with structural similarity. Experiments on samples showed that naïve and brolucizumab-treated ADA-positive patients developed a class-switched, high-affinity immune response, with several linear epitopes being recognized by ADAs. Only patients with RV/RO showed a meaningful T cell response upon recall with brolucizumab. Further studies in cynomolgus monkeys preimmunized against brolucizumab with adjuvant followed by intravitreal brolucizumab challenge demonstrated that high ADA titers were required to generate ocular inflammation and vasculitis/vascular thrombosis, comparable to RV/RO in humans. Immunogenicity therefore seems to be a prerequisite to develop RV/RO. However, because only 2.1% of patients with ADA develop RV/RO, additional factors must play a role in the development of RV/RO.


Subject(s)
Retinal Vasculitis , Animals , Humans , Adjuvants, Immunologic , Angiogenesis Inhibitors , Inflammation , Intravitreal Injections , Macaca fascicularis , Vascular Endothelial Growth Factor A
2.
Sci Transl Med ; 15(681): eabq5068, 2023 02.
Article in English | MEDLINE | ID: mdl-36724241

ABSTRACT

Immunogenicity against intravitreally administered brolucizumab has been previously described and associated with cases of severe intraocular inflammation, including retinal vasculitis/retinal vascular occlusion (RV/RO). The presence of antidrug antibodies (ADAs) in these patients led to the initial hypothesis that immune complexes could be key mediators. Although the formation of ADAs and immune complexes may be a prerequisite, other factors likely contribute to some patients having RV/RO, whereas the vast majority do not. To identify and characterize the mechanistic drivers underlying the immunogenicity of brolucizumab and the consequence of subsequent ADA-induced immune complex formation, a translational approach was performed to bridge physicochemical characterization, structural modeling, sequence analysis, immunological assays, and a quantitative systems pharmacology model that mimics physiological conditions within the eye. This approach revealed that multiple factors contributed to the increased immunogenic potential of brolucizumab, including a linear epitope shared with bacteria, non-natural surfaces due to the single-chain variable fragment format, and non-native drug species that may form over prolonged time in the eye. Consideration of intraocular drug pharmacology and disease state in a quantitative systems pharmacology model suggested that immune complexes could form at immunologically relevant concentrations modulated by dose intensity. Assays using circulating immune cells from treated patients or treatment-naïve healthy volunteers revealed the capacity of immune complexes to trigger cellular responses such as enhanced antigen presentation, platelet aggregation, endothelial cell activation, and cytokine release. Together, these studies informed a mechanistic understanding of the clinically observed immunogenicity of brolucizumab and associated cases of RV/RO.


Subject(s)
Antigen-Antibody Complex , Root Cause Analysis , Humans , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Inflammation , Angiogenesis Inhibitors , Intravitreal Injections
3.
JCI Insight ; 7(13)2022 07 08.
Article in English | MEDLINE | ID: mdl-35801586

ABSTRACT

IL-1 receptor-activated kinase 1 (IRAK1) is involved in signal transduction downstream of many TLRs and the IL-1R. Its potential as a drug target for chronic inflammatory diseases is underappreciated. To study its functional role in joint inflammation, we generated a mouse model expressing a functionally inactive IRAK1 (IRAK1 kinase deficient, IRAK1KD), which also displayed reduced IRAK1 protein expression and cell type-specific deficiencies of TLR signaling. The serum transfer model of arthritis revealed a potentially novel role of IRAK1 for disease development and neutrophil chemoattraction exclusively via its activity in nonhematopoietic cells. Consistently, IRAK1KD synovial fibroblasts showed reduced secretion of neutrophil chemoattractant chemokines following stimulation with IL-1ß or human synovial fluids from patients with rheumatoid arthritis (RA) and gout. Together with patients with RA showing prominent IRAK1 expression in fibroblasts of the synovial lining, these data suggest that targeting IRAK1 may be therapeutically beneficial. As pharmacological inhibition of IRAK1 kinase activity had only mild effects on synovial fibroblasts from mice and patients with RA, targeted degradation of IRAK1 may be the preferred pharmacologic modality. Collectively, these data position IRAK1 as a central regulator of the IL-1ß-dependent local inflammatory milieu of the joints and a potential therapeutic target for inflammatory arthritis.


Subject(s)
Arthritis, Rheumatoid , Interleukin-1 Receptor-Associated Kinases , Neutrophils , Synovial Membrane , Animals , Arthritis, Rheumatoid/metabolism , Cells, Cultured , Disease Models, Animal , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-8/metabolism , Mice , Neutrophils/metabolism , Synovial Membrane/metabolism
4.
Cell Rep ; 37(6): 109977, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34758308

ABSTRACT

Tumor necrosis factor (TNF) is a key driver of several inflammatory diseases, such as rheumatoid arthritis, inflammatory bowel disease, and psoriasis, in which affected tissues show an interferon-stimulated gene signature. Here, we demonstrate that TNF triggers a type-I interferon response that is dependent on the cyclic guanosine monophosphate-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. We show that TNF inhibits PINK1-mediated mitophagy and leads to altered mitochondrial function and to an increase in cytosolic mtDNA levels. Using cGAS-chromatin immunoprecipitation (ChIP), we demonstrate that cytosolic mtDNA binds to cGAS after TNF treatment. Furthermore, TNF induces a cGAS-STING-dependent transcriptional response that mimics that of macrophages from rheumatoid arthritis patients. Finally, in an inflammatory arthritis mouse model, cGAS deficiency blocked interferon responses and reduced inflammatory cell infiltration and joint swelling. These findings elucidate a molecular mechanism linking TNF to type-I interferon signaling and suggest a potential benefit for therapeutic targeting of cGAS/STING in TNF-driven diseases.


Subject(s)
Arthritis, Experimental/immunology , DNA, Mitochondrial/metabolism , Immunity, Innate , Inflammation/immunology , Interferon Type I/pharmacology , Membrane Proteins/metabolism , Nucleotidyltransferases/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , DNA, Mitochondrial/drug effects , Female , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Macrophages/immunology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitophagy
5.
J Med Chem ; 64(4): 1889-1903, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33592148

ABSTRACT

The cytosolic metalloenzyme leukotriene A4 hydrolase (LTA4H) is the final and rate-limiting enzyme in the biosynthesis of pro-inflammatory leukotriene B4 (LTB4). Preclinical studies have validated this enzyme as an attractive drug target in chronic inflammatory diseases. Despite several attempts, no LTA4H inhibitor has reached the market, yet. Herein, we disclose the discovery and preclinical profile of LYS006, a highly potent and selective LTA4H inhibitor. A focused fragment screen identified hits that could be cocrystallized with LTA4H and inspired a fragment merging. Further optimization led to chiral amino acids and ultimately to LYS006, a picomolar LTA4H inhibitor with exquisite whole blood potency and long-lasting pharmacodynamic effects. Due to its high selectivity and its ability to fully suppress LTB4 generation at low exposures in vivo, LYS006 has the potential for a best-in-class LTA4H inhibitor and is currently investigated in phase II clinical trials in inflammatory acne, hidradenitis suppurativa, ulcerative colitis, and NASH.


Subject(s)
Aminobutyrates/therapeutic use , Anti-Inflammatory Agents/pharmacology , Enzyme Inhibitors/therapeutic use , Epoxide Hydrolases/antagonists & inhibitors , Pyridines/therapeutic use , Aminobutyrates/chemical synthesis , Aminobutyrates/pharmacokinetics , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacokinetics , Arthritis, Experimental/drug therapy , Dogs , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Inflammation/drug therapy , Male , Mice, Inbred C57BL , Molecular Structure , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Rats, Wistar , Structure-Activity Relationship
6.
J Med Chem ; 63(17): 9856-9875, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32856916

ABSTRACT

G-protein-coupled receptor SUCNR1 (succinate receptor 1 or GPR91) senses the citric cycle intermediate succinate and is implicated in various pathological conditions such as rheumatoid arthritis, liver fibrosis, or obesity. Here, we describe a novel SUCNR1 antagonist scaffold discovered by high-throughput screening. The poor permeation and absorption properties of the most potent compounds, which were zwitterionic in nature, could be improved by the formation of an internal salt bridge, which helped in shielding the two opposite charges and thus also the high polarity of zwitterions with separated charges. The designed compounds containing such a salt bridge reached high oral bioavailability and oral exposure. We believe that this principle could find a broad interest in the medicinal chemistry field as it can be useful not only for the modulation of properties in zwitterionic compounds but also in acidic or basic compounds with poor permeation.


Subject(s)
Benzamides/pharmacology , Phenylacetates/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Benzamides/chemical synthesis , Benzamides/metabolism , Benzamides/pharmacokinetics , Cell Line , Drug Discovery , Humans , Male , Mice, Inbred C57BL , Phenylacetates/chemical synthesis , Phenylacetates/metabolism , Phenylacetates/pharmacokinetics , Protein Binding , Rats , Receptors, G-Protein-Coupled/metabolism , Static Electricity
7.
J Control Release ; 326: 172-180, 2020 10 10.
Article in English | MEDLINE | ID: mdl-32653504

ABSTRACT

Anti-inflammatory cytokines are a promising class of therapeutics for treatment of rheumatoid arthritis (RA), but their use is currently limited by a rapid clearance and systemic toxicity. Interleukin-4 is a small cytokine with potential for RA therapy. To increase its pharmacokinetic features, we engineered a murine IL4 conjugate by incorporating an unnatural amino acid through genetic code expansion to which PEG-folate, as a targeting moiety and PEG alone as control, were site-specifically bound. Both IL4 conjugates retained bioactivity and induced primary murine macrophage polarization into an alternatively activated (M2) related phenotype. The PEGylated conjugates had a terminal half-life of about four hours in healthy mice compared to unPEGylated IL4 (0.76 h). We showed that both conjugates successfully accumulated into arthritic joints in an antigen-induced arthritis (AIA) mouse model, as assessed by non-invasive fluorescence imaging. The modular nature of the IL4 conjugate chemistry presented herein facilitates easy adaption of PEG chain length and targeting moieties for further improvement of half-life and targeting function for future efficacy studies.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Interleukin-4/therapeutic use , Amino Acids , Animals , Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Interleukin-4/administration & dosage , Mice , Polyethylene Glycols
8.
Front Immunol ; 11: 745, 2020.
Article in English | MEDLINE | ID: mdl-32425939

ABSTRACT

Genetic disruption or short-term pharmacological inhibition of MALT1 protease is effective in several preclinical models of autoimmunity and B cell malignancies. Despite these protective effects, the severe reduction in regulatory T cells (Tregs) and the associated IPEX-like pathology occurring upon congenital disruption of the MALT1 protease in mice has raised concerns about the long-term safety of MALT1 inhibition. Here we describe the results of a series of toxicology studies in rat and dog species using MLT-943, a novel potent and selective MALT1 protease inhibitor. While MLT-943 effectively prevented T cell-dependent B cell immune responses and reduced joint inflammation in the collagen-induced arthritis rat pharmacology model, in both preclinical species, pharmacological inhibition of MALT1 was associated with a rapid and dose-dependent reduction in Tregs and resulted in the progressive appearance of immune abnormalities and clinical signs of an IPEX-like pathology. At the 13-week time point, rats displayed severe intestinal inflammation associated with mast cell activation, high serum IgE levels, systemic T cell activation and mononuclear cell infiltration in multiple tissues. Importantly, using thymectomized rats we demonstrated that MALT1 protease inhibition affects peripheral Treg frequency independently of effects on thymic Treg output and development. Our data confirm the therapeutic potential of MALT1 protease inhibitors but highlight the safety risks and challenges to consider before potential application of such inhibitors into the clinic.


Subject(s)
Diabetes Mellitus, Type 1/congenital , Diarrhea/etiology , Genetic Diseases, X-Linked/etiology , Immune System Diseases/congenital , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , Animals , Diabetes Mellitus, Type 1/etiology , Dogs , Female , Humans , Immune System Diseases/etiology , Inflammation/chemically induced , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred Lew , Rats, Wistar , T-Lymphocytes, Regulatory/immunology
9.
J Med Chem ; 63(10): 5102-5118, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32083858

ABSTRACT

Bruton's tyrosine kinase (BTK), a cytoplasmic tyrosine kinase, plays a central role in immunity and is considered an attractive target for treating autoimmune diseases. The use of currently marketed covalent BTK inhibitors is limited to oncology indications based on their suboptimal kinase selectivity. We describe the discovery and preclinical profile of LOU064 (remibrutinib, 25), a potent, highly selective covalent BTK inhibitor. LOU064 exhibits an exquisite kinase selectivity due to binding to an inactive conformation of BTK and has the potential for a best-in-class covalent BTK inhibitor for the treatment of autoimmune diseases. It demonstrates potent in vivo target occupancy with an EC90 of 1.6 mg/kg and dose-dependent efficacy in rat collagen-induced arthritis. LOU064 is currently being tested in phase 2 clinical studies for chronic spontaneous urticaria and Sjoegren's syndrome.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/metabolism , Drug Discovery/methods , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Agammaglobulinaemia Tyrosine Kinase/chemistry , Animals , Benzamides/chemistry , Benzamides/metabolism , Benzamides/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Crystallography, X-Ray/methods , Dogs , Dose-Response Relationship, Drug , Female , Humans , Mice , Protein Binding/physiology , Protein Kinase Inhibitors/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Rats, Inbred Lew , Sheep
10.
Eur Respir J ; 55(3)2020 03.
Article in English | MEDLINE | ID: mdl-31949113

ABSTRACT

Sarcoidosis is a disease characterised by granuloma formation. There is an unmet need for new treatment strategies beyond corticosteroids. The NLRP3 inflammasome pathway is expressed in innate immune cells and senses danger signals to elicit inflammatory interleukin (IL)-1ß; it has recently become a druggable target. This prompted us to test the role of the NLRP3 inflammasome and IL-1ß pathway in granuloma formation and sarcoidosis.19 sarcoid patients and 19 healthy volunteers were recruited into this pilot study. NLRP3 inflammasome activity was measured in bronchoalveolar lavage (BAL) cells and lung and skin biopsies using immunohistochemistry, Western blot, reverse-transcriptase PCR and ELISA. For in vivo experiments we used the trehalose 6,6'-dimycolate-granuloma mouse model and evaluated lung granuloma burden in miR-223 knockout and NLRP3 knockout mice, as well as the treatment effects of MCC950 and anti-IL-1ß antibody therapy.We found strong upregulation of the NLRP3 inflammasome pathway, evidenced by expression of activated NLRP3 inflammasome components, including cleaved caspase-1 and IL-1ß in lung granuloma, and increased IL-1ß release of BAL cells from sarcoid patients compared to healthy volunteers (p=0.006). mRNA levels of miR-223, a micro-RNA downregulating NLRP3, were decreased and NLRP3 mRNA correspondingly increased in alveolar macrophages from sarcoid patients (p<0.005). NLRP3 knockout mice showed decreased and miR-223 knockout mice increased granuloma formation compared to wild-type mice. Pharmacological interference using NLRP3 pathway inhibitor MCC950 or an anti-IL-1ß antibody resulted in reduced granuloma formation (p<0.02).In conclusion, our data provide evidence of upregulated inflammasome and IL-1ß pathway activation in sarcoidosis and suggest both as valid therapeutic targets.


Subject(s)
Granuloma , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Sarcoidosis , Animals , Caspase 1 , Humans , Interleukin-1beta , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pilot Projects
11.
Arthritis Rheumatol ; 72(6): 919-930, 2020 06.
Article in English | MEDLINE | ID: mdl-31943941

ABSTRACT

OBJECTIVE: Fcγ receptors (FcγR) play important roles in both protective and pathogenic immune responses. The assembly of the CBM signalosome encompassing caspase recruitment domain-containing protein 9, B cell CLL/lymphoma 10, and mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT-1) is required for optimal FcγR-induced canonical NF-κB activation and proinflammatory cytokine release. This study was undertaken to clarify the relevance of MALT-1 protease activity in FcγR-driven events and evaluate the therapeutic potential of selective MALT-1 protease inhibitors in FcγR-mediated diseases. METHODS: Using genetic and pharmacologic disruption of MALT-1 scaffolding and enzymatic activity, we assessed the relevance of MALT-1 function in murine and human primary myeloid cells upon stimulation with immune complexes (ICs) and in murine models of autoantibody-driven arthritis and immune thrombocytopenic purpura (ITP). RESULTS: MALT-1 protease function is essential for optimal FcγR-induced production of proinflammatory cytokines by various murine and human myeloid cells stimulated with ICs. In contrast, MALT-1 protease inhibition did not affect the Syk-dependent, FcγR-mediated production of reactive oxygen species or leukotriene B4 . Notably, pharmacologic MALT-1 protease inhibition in vivo reduced joint inflammation in the murine K/BxN serum-induced arthritis model (mean area under the curve for paw swelling of 45.42% versus 100% in control mice; P = 0.0007) but did not affect platelet depletion in a passive model of ITP. CONCLUSION: Our findings indicate a specific contribution of MALT-1 protease activity to FcγR-mediated events and suggest that MALT-1 protease inhibitors have therapeutic potential in a subset of FcγR-driven inflammatory disorders.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/immunology , Receptors, IgG/immunology , Animals , Antigen-Antibody Complex/metabolism , Blood Platelets/metabolism , Cytokines/immunology , Disease Models, Animal , Humans , Mice , Myeloid Cells/metabolism
12.
Nature ; 574(7779): 581-585, 2019 10.
Article in English | MEDLINE | ID: mdl-31645725

ABSTRACT

The tricarboxylic acid cycle intermediate succinate is involved in metabolic processes and plays a crucial role in the homeostasis of mitochondrial reactive oxygen species1. The receptor responsible for succinate signalling, SUCNR1 (also known as GPR91), is a member of the G-protein-coupled-receptor family2 and links succinate signalling to renin-induced hypertension, retinal angiogenesis and inflammation3-5. Because SUCNR1 senses succinate as an immunological danger signal6-which has relevance for diseases including ulcerative colitis, liver fibrosis7, diabetes and rheumatoid arthritis3,8-it is of interest as a therapeutic target. Here we report the high-resolution crystal structure of rat SUCNR1 in complex with an intracellular binding nanobody in the inactive conformation. Structure-based mutagenesis and radioligand-binding studies, in conjunction with molecular modelling, identified key residues for species-selective antagonist binding and enabled the determination of the high-resolution crystal structure of a humanized rat SUCNR1 in complex with a high-affinity, human-selective antagonist denoted NF-56-EJ40. We anticipate that these structural insights into the architecture of the succinate receptor and its antagonist selectivity will enable structure-based drug discovery and will further help to elucidate the function of SUCNR1 in vitro and in vivo.


Subject(s)
Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Piperazines/chemistry , Piperazines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Animals , Apoproteins/antagonists & inhibitors , Apoproteins/chemistry , Apoproteins/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Rats , Receptors, G-Protein-Coupled/metabolism , Receptors, Purinergic P2Y1/chemistry , Signal Transduction , Single-Domain Antibodies/chemistry , Species Specificity , Succinic Acid/metabolism
13.
Proc Natl Acad Sci U S A ; 116(16): 7926-7931, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30926668

ABSTRACT

Dysregulation of the alternative complement pathway (AP) predisposes individuals to a number of diseases including paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, and C3 glomerulopathy. Moreover, glomerular Ig deposits can lead to complement-driven nephropathies. Here we describe the discovery of a highly potent, reversible, and selective small-molecule inhibitor of factor B, a serine protease that drives the central amplification loop of the AP. Oral administration of the inhibitor prevents KRN-induced arthritis in mice and is effective upon prophylactic and therapeutic dosing in an experimental model of membranous nephropathy in rats. In addition, inhibition of factor B prevents complement activation in sera from C3 glomerulopathy patients and the hemolysis of human PNH erythrocytes. These data demonstrate the potential therapeutic value of using a factor B inhibitor for systemic treatment of complement-mediated diseases and provide a basis for its clinical development.


Subject(s)
Complement Factor B/antagonists & inhibitors , Complement Pathway, Alternative/drug effects , Drug Discovery/methods , Immunologic Factors/pharmacology , Animals , Disease Models, Animal , Glomerulonephritis, Membranous/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Rats, Sprague-Dawley
14.
ACS Med Chem Lett ; 8(9): 975-980, 2017 Sep 14.
Article in English | MEDLINE | ID: mdl-28947947

ABSTRACT

The predominant expression of phosphoinositide 3-kinase δ (PI3Kδ) in leukocytes and its critical role in B and T cell functions led to the hypothesis that selective inhibitors of this isoform would have potential as therapeutics for the treatment of allergic and inflammatory disease. Targeting specifically PI3Kδ should avoid potential side effects associated with the ubiquitously expressed PI3Kα and ß isoforms. We disclose how morphing the heterocyclic core of previously discovered 4,6-diaryl quinazolines to a significantly less lipophilic 5,6,7,8-tetrahydropyrido[4,3-d]pyrimidine, followed by replacement of one of the phenyl groups with a pyrrolidine-3-amine, led to a compound series with an optimal on-target profile and good ADME properties. A final lipophilicity adjustment led to the discovery of CDZ173 (leniolisib), a potent PI3Kδ selective inhibitor with suitable properties and efficacy for clinical development as an anti-inflammatory therapeutic. In vitro, CDZ173 inhibits a large spectrum of immune cell functions, as demonstrated in B and T cells, neutrophils, monocytes, basophils, plasmocytoid dendritic cells, and mast cells. In vivo, CDZ173 inhibits B cell activation in rats and monkeys in a concentration- and time-dependent manner. After prophylactic or therapeutic dosing, CDZ173 potently inhibited antigen-specific antibody production and reduced disease symptoms in a rat collagen-induced arthritis model. Structurally, CDZ173 differs significantly from the first generation of PI3Kδ and PI3Kγδ-selective clinical compounds. Therefore, CDZ173 could differentiate by a more favorable safety profile. CDZ173 is currently in clinical studies in patients suffering from primary Sjögren's syndrome and in APDS/PASLI, a disease caused by gain-of-function mutations of PI3Kδ.

15.
Bioorg Med Chem ; 25(16): 4512-4525, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28689977

ABSTRACT

GPR4, a G-protein coupled receptor, functions as a proton sensor being activated by extracellular acidic pH and has been implicated in playing a key role in acidosis associated with a variety of inflammatory conditions. An orally active GPR4 antagonist 39c was developed, starting from a high throughput screening hit 1. The compound shows potent cellular activity and is efficacious in animal models of angiogenesis, inflammation and pain.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Drug Design , Inflammation/drug therapy , Receptors, G-Protein-Coupled/antagonists & inhibitors , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Arthritis/drug therapy , Arthritis/metabolism , COS Cells , Chlorocebus aethiops , Dose-Response Relationship, Drug , Female , HEK293 Cells , HeLa Cells , Humans , Inflammation/metabolism , Mice , Molecular Structure , Pain/drug therapy , Pain/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship
16.
J Med Chem ; 60(9): 3672-3683, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28445047

ABSTRACT

A novel, selective, and efficacious GPR4 antagonist 13 was developed starting from lead compound 1a. While compound 1a showed promising efficacy in several disease models, its binding to a H3 receptor as well as a hERG channel prevented it from further development. Therefore, a new round of optimization addressing the key liabilities was performed and led to discovery of compound 13 with an improved profile. Compound 13 showed significant efficacy in the rat antigen induced arthritis as well as in the hyperalgesia and angiogenesis model at a well-tolerated dose of 30 mg/kg.


Subject(s)
Inflammation/prevention & control , Neovascularization, Physiologic/drug effects , Nociception/drug effects , Receptors, G-Protein-Coupled/antagonists & inhibitors , Administration, Oral , Animals , Drug Design , Female , HEK293 Cells , Humans , Rats , Rats, Sprague-Dawley , Receptors, Histamine H3/metabolism
17.
J Exp Med ; 213(9): 1655-62, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27481132

ABSTRACT

When SUCNR1/GPR91-expressing macrophages are activated by inflammatory signals, they change their metabolism and accumulate succinate. In this study, we show that during this activation, macrophages release succinate into the extracellular milieu. They simultaneously up-regulate GPR91, which functions as an autocrine and paracrine sensor for extracellular succinate to enhance IL-1ß production. GPR91-deficient mice lack this metabolic sensor and show reduced macrophage activation and production of IL-1ß during antigen-induced arthritis. Succinate is abundant in synovial fluids from rheumatoid arthritis (RA) patients, and these fluids elicit IL-1ß release from macrophages in a GPR91-dependent manner. Together, we reveal a GPR91/succinate-dependent feed-forward loop of macrophage activation and propose GPR91 antagonists as novel therapeutic principles to treat RA.


Subject(s)
Arthritis, Rheumatoid/etiology , Macrophages/metabolism , Receptors, G-Protein-Coupled/physiology , Succinic Acid/metabolism , Animals , Arthritis, Rheumatoid/metabolism , Female , Humans , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/antagonists & inhibitors , U937 Cells
18.
J Med Chem ; 59(1): 132-46, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26629594

ABSTRACT

This paper describes the identification of 6-(pyrimidin-4-yloxy)-naphthalene-1-carboxamides as a new class of potent and selective human vascular endothelial growth factor receptor 2 (VEGFR2) tyrosine kinase inhibitors. In biochemical and cellular assays, the compounds exhibit single-digit nanomolar potency toward VEGFR2. Compounds of this series show good exposure in rodents when dosed orally. They potently inhibit VEGF-driven angiogenesis in a chamber model and rodent tumor models at daily doses of less than 3 mg/kg by targeting the tumor vasculature as demonstrated by ELISA for TIE-2 in lysates or by immunohistochemical analysis. This novel series of compounds shows a potential for the treatment of solid tumors and other diseases where angiogenesis plays an important role.


Subject(s)
Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacokinetics , Animals , CHO Cells , Cell Proliferation/drug effects , Cricetinae , Cricetulus , Female , Human Umbilical Vein Endothelial Cells , Humans , Melanoma, Experimental/drug therapy , Mice , Models, Molecular , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Phosphorylation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Vascular Endothelial Growth Factor A/pharmacology , Xenograft Model Antitumor Assays
19.
J Med Chem ; 58(4): 1950-63, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25633741

ABSTRACT

We describe the discovery of selective and potent Syk inhibitor 11, which exhibited favorable PK profiles in rat and dog and was found to be active in a collagen-induced arthritis model in rats. Compound 11 was selected for further profiling, but, unfortunately, in GLP toxicological studies it showed liver findings in rat and dog. Nevertheless, 11 could become a valuable tool compound to investigate the rich biology of Syk in vitro and in vivo.


Subject(s)
Arthritis, Experimental/drug therapy , Drug Discovery , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Arthritis, Experimental/chemically induced , Collagen , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Female , Humans , Intracellular Signaling Peptides and Proteins/blood , Intracellular Signaling Peptides and Proteins/metabolism , Liver/drug effects , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Conformation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/blood , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Inbred Lew , Structure-Activity Relationship , Syk Kinase
20.
Chembiochem ; 14(18): 2512-8, 2013 Dec 16.
Article in English | MEDLINE | ID: mdl-24222525

ABSTRACT

Cellular behaviors are governed by combinations of systemic and microenvironmental factors; together, these regulate cell signaling responses to growth factors. This contextual microenvironmental influence also determines drug sensitivity. Hence using in vitro systems that model contextual cellular behavior is highly beneficial for effective therapeutic development. Angiogenesis (formation of blood vessels) is driven by a series of dynamic endothelial cell signaling responses to growth factors under the influence of the vascular extracellular matrix and adjacent pericytes. In vitro primary human vascular cell co-cultures self-assemble into capillary-like structures through reciprocal heterotypic interactions that mimic angiogenic context dynamics. By using temporal live-cell imaging-based analysis, unique angiogenic microenvironments can be delineated to quantify the contextual activity of compound inhibitors. We used this in vitro organotypic contextual screening approach to conduct structure-activity relationship analysis on a combretastatin A-4 analogue series to identify novel compounds with potent vascular disrupting activity in vivo.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Drug Evaluation, Preclinical/methods , Angiogenesis Inhibitors/chemistry , Animals , Cell Line , Coculture Techniques/methods , Human Umbilical Vein Endothelial Cells , Humans , Pulmonary Artery/cytology , Structure-Activity Relationship , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...