Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Exp Dermatol ; 33(5): e15099, 2024 05.
Article in English | MEDLINE | ID: mdl-38794814

ABSTRACT

Suitable human models for the development and characterization of topical compounds for inflammatory skin diseases such as atopic dermatitis are not readily available to date. We describe here the development of a translational model involving healthy human skin mimicking major aspects of AD and its application for the characterization of topical Janus kinase inhibitors. Full thickness human abdominal skin obtained from plastic surgery stimulated in vitro with IL4 and IL13 shows molecular features of AD. This is evidenced by STAT6 phosphorylation assessed by immunohistochemistry and analysis of skin lysates. Broad transcriptome changes assessed by AmpliSeq followed by gene set variation analysis showed a consistent upregulation of gene signatures characterizing AD in this model. Topical application of experimental formulations of compounds targeting the JAK pathway to full thickness skin normalizes the molecular features of AD induced by IL4 and IL13 stimulation. The inhibitory effects of topical JAK inhibitors on molecular features of AD are supported by pharmacokinetic analysis. The model described here is suited for the characterization of topical compounds for AD and has the potential to be extended to other inflammatory skin diseases and pathophysiological pathways.


Subject(s)
Dermatitis, Atopic , Janus Kinase Inhibitors , Skin , Humans , Dermatitis, Atopic/drug therapy , Skin/metabolism , Skin/drug effects , Janus Kinase Inhibitors/pharmacology , STAT6 Transcription Factor/metabolism , Interleukin-4/metabolism , Interleukin-13/metabolism , Phosphorylation , Transcriptome , Models, Biological , Pyrimidines/pharmacology , Administration, Topical , Piperidines
2.
J Med Chem ; 67(6): 5093-5108, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38476002

ABSTRACT

Leukotriene A4 hydrolase (LTA4H) is the final and rate-limiting enzyme in the biosynthesis of pro-inflammatory leukotriene B4 (LTB4). Preclinical studies have provided strong evidence that LTA4H is an attractive drug target for the treatment of chronic inflammatory diseases. Here, we describe the transformation of compound 2, a fragment-like hit, into the potent inhibitor of LTA4H 3. Our strategy involved two key steps. First, we aimed to increase the polarity of fragment 2 to improve its drug-likeness, particularly its solubility, while preserving both its promising potency and low molecular weight. Second, we utilized structural information and incorporated a basic amino function, which allowed for the formation of an essential hydrogen bond with Q136 of LTA4H and consequently enhanced the potency. Compound 3 exhibited exceptional selectivity and showed oral efficacy in a KRN passive serum-induced arthritis model in mice. The anticipated human dose to achieve 90% target engagement at the trough concentration was determined to be 40 mg administered once daily.


Subject(s)
Enzyme Inhibitors , Epoxide Hydrolases , Mice , Humans , Animals , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Leukotriene B4
3.
Clin Transl Sci ; 17(2): e13724, 2024 02.
Article in English | MEDLINE | ID: mdl-38407540

ABSTRACT

LYS006 is a novel, highly potent and selective, new-generation leukotriene A4 hydrolase (LTA4H) inhibitor in clinical development for the treatment of neutrophil-driven inflammatory diseases. We describe the complex pharmacokinetic to pharmacodynamic (PD) relationship in blood, plasma, and skin of LYS006-treated nonclinical species and healthy human participants. In a randomized first in human study, participants were exposed to single ascending doses up to 100 mg and multiple ascending doses up to 80 mg b.i.d.. LYS006 showed rapid absorption, overall dose proportional plasma exposure and nonlinear blood to plasma distribution caused by saturable target binding. The compound efficiently inhibited LTB4 production in human blood and skin blister cells, leading to greater than 90% predose target inhibition from day 1 after treatment initiation at doses of 20 mg b.i.d. and above. Slow re-distribution from target expressing cells resulted in a long terminal half-life and a long-lasting PD effect in ex vivo stimulated blood and skin cells despite low plasma exposures. LYS006 was well-tolerated and demonstrated a favorable safety profile up to highest doses tested, without any dose-limiting toxicity. This supported further clinical development in phase II studies in predominantly neutrophil-driven inflammatory conditions, such as hidradenitis suppurativa, inflammatory acne, and ulcerative colitis.


Subject(s)
Epoxide Hydrolases , Plasma , Humans , Neutrophils , Skin
4.
J Med Chem ; 66(23): 16410-16425, 2023 12 14.
Article in English | MEDLINE | ID: mdl-38015154

ABSTRACT

The discovery of chiral amino alcohols derived from our previously disclosed clinical LTA4H inhibitor LYS006 is described. In a biochemical assay, their optical antipodes showed similar potencies, which could be rationalized by the cocrystal structures of these compounds bound to LTA4H. Despite comparable stabilities in liver microsomes, they showed distinct in vivo PK properties. Selective O-phosphorylation of the (R)-enantiomers in blood led to clearance values above the hepatic blood flow, whereas the (S)-enantiomers were unaffected and exhibited satisfactory metabolic stabilities in vivo. Introduction of two pyrazole rings led to compound (S)-2 with a more balanced distribution of polarity across the molecule, exhibiting high selectivity and excellent potency in vitro and in vivo. Furthermore, compound (S)-2 showed favorable profiles in 16-week IND-enabling toxicology studies in dogs and rats. Based on allometric scaling and potency in whole blood, compound (S)-2 has the potential for a low oral efficacious dose administered once daily.


Subject(s)
Epoxide Hydrolases , Liver , Rats , Animals , Dogs , Epoxide Hydrolases/metabolism , Liver/metabolism , Microsomes, Liver/metabolism
5.
J Med Chem ; 66(21): 15042-15053, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37906573

ABSTRACT

We describe the discovery and characterization of the supersoft topical JAK inhibitor 3(R), which is potent in biochemical and cellular assays as well as in human skin models. In blood, the neutral ester 3(R) is rapidly hydrolyzed (t1/2 ∼ 6 min) to the corresponding charged carboxylic acid 4 exhibiting >30-fold reduced permeability. Consequently, acid 4 does not reach the intracellular JAK kinases and is inactive in cellular assays and in blood. Thus, hydrolysis by blood esterases leads to the rapid deactivation of topically active ester 3(R) at a rate beyond the maximal hepatic clearance.


Subject(s)
Janus Kinase Inhibitors , Humans , Skin , Esterases , Hydrolysis , Esters
6.
ACS Med Chem Lett ; 14(6): 841-845, 2023 Jun 08.
Article in English | MEDLINE | ID: mdl-37312861

ABSTRACT

We present a novel concept for the design of supersoft topical drugs. Enzymatic cleavage of the carbonate ester of the potent pan Janus kinase (JAK) inhibitor 2 releases hydroxypyridine 3. Due to hydroxypyridine-pyridone tautomerism, 3 undergoes a rapid conformational change preventing the compound to assume the bioactive conformation required for binding to JAK kinases. We demonstrate that the hydrolysis in human blood and the subsequent shape change lead to the deactivation of 2.

7.
J Med Chem ; 66(3): 2161-2168, 2023 02 09.
Article in English | MEDLINE | ID: mdl-36657024

ABSTRACT

The JAK kinases JAK1, JAK2, JAK3, and TYK2 play key roles in cytokine signaling. Activation of the JAK/STAT pathways is linked to many diseases involving the immune system, including atopic dermatitis. As systemic JAK inhibitor pharmacology is associated with side effects, topical administration to the skin has been considered to locally restrict the site of action. Several orally bioavailable JAK inhibitors repurposed for topical use have been recently approved or are in clinical development. Here, we disclose our clinical candidate CEE321, which is a potent pan JAK inhibitor in enzyme and cellular assays. In contrast to repurposed oral drugs, CEE321 does not display high potency in blood and has a high clearance in vivo. Therefore, we consider CEE321 to be a "soft drug". When applied topically to human skin that was stimulated with the cytokines IL4 and IL13 ex vivo, CEE321 potently inhibited biomarkers relevant to atopic dermatitis.


Subject(s)
Dermatitis, Atopic , Janus Kinase Inhibitors , Humans , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/metabolism , Dermatitis, Atopic/drug therapy , Janus Kinases , Skin/metabolism , Cytokines/metabolism
8.
Expert Opin Drug Discov ; 16(12): 1483-1495, 2021 12.
Article in English | MEDLINE | ID: mdl-34191664

ABSTRACT

IntroductionLeukotriene A4 hydrolase (LTA4H) is the final and rate limiting enzyme regulating the biosynthesis of leukotriene B4 (LTB4), a pro-inflammatory lipid mediator implicated in a large number of inflammatory pathologies. Inhibition of LTA4H not only prevents LTB4 biosynthesis but also induces a lipid mediator class-switch within the 5-lipoxygenase pathway, elevating biosynthesis of the anti-inflammatory lipid mediator Lipoxin A4. Ample preclinical evidence advocates LTA4H as attractive drug target for the treatment of chronic inflammatory diseases.Areas coveredThis review covers details about the biochemistry of LTA4H and describes its role in regulating pro- and anti-inflammatory mediator generation. It summarizes recent efforts in medicinal chemistry toward novel LTA4H inhibitors, recent clinical trials testing LTA4H inhibitors in pulmonary inflammatory diseases, and potential reasons for the discontinuation of former development programs.Expert opinionGiven the prominent role of LTB4 in initiating and perpetuating inflammation, LTA4H remains an appealing drug target. The reason former attempts targeting this enzyme have not met with success in the clinic can be attributed to compound-specific liabilities of first-generation inhibitors and/or choice of target indications to test this mode of action. A new generation of highly potent and selective LTA4H inhibitors is currently undergoing clinical testing in indications with a strong link to LTB4 biology.


Subject(s)
Enzyme Inhibitors , Epoxide Hydrolases , Anti-Inflammatory Agents/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/metabolism , Humans
9.
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
10.
Bioorg Med Chem Lett ; 25(20): 4642-7, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26320624

ABSTRACT

Design and optimization of benzo- and pyrido-thiazoles/isothiazoles are reported leading to the discovery of the potent, orally bioavailable Syk inhibitor 5, which was found to be active in a rat PK/PD model. Compound 5 showed acceptable overall kinase selectivity. However, in addition to Syk it also inhibited Aurora kinase in enzymatic and cellular settings leading to findings in the micronucleus assay. As a consequence, compound 5 was not further pursued.


Subject(s)
Disease Models, Animal , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Thiazoles/pharmacology , Administration, Oral , Animals , Biological Availability , Dose-Response Relationship, Drug , Intracellular Signaling Peptides and Proteins/metabolism , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Structure-Activity Relationship , Syk Kinase , Thiazoles/administration & dosage , Thiazoles/chemistry
11.
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
12.
Bioorg Med Chem Lett ; 24(19): 4617-4621, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25217444

ABSTRACT

Jak3, together with Jak1, is involved in signal transduction initiated by cytokines signaling through the common gamma chain which are important in immune homeostasis and immune pathologies. Based on genetic evidence Jak3 has been considered to be an attractive target for immunosuppression. The Jak inhibitor tofacitinib (CP-690,550) which is an approved drug for rheumatoid arthritis was originally introduced as a selective Jak3 inhibitor, however, it also inhibits Jak1 and Jak2. The search for new selective Jak3 inhibitors has yielded several compounds whose profiles will be reviewed here. Implications on Jak3 as a therapeutic target are also discussed.


Subject(s)
Janus Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Humans , Janus Kinase 3/metabolism , Molecular Structure , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
13.
Bioorg Med Chem Lett ; 24(10): 2278-82, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24726806

ABSTRACT

We describe two series of Syk inhibitors which potently abrogate Syk kinase function in enzymatic assays, cellular assays and in primary cells in the presence of blood. Introduction of a 7-aminoindole substituent led to derivatives with good kinase selectivity and little or no hERG channel inhibition (3b, 10c).


Subject(s)
Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/blood , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/blood , Humans , Indoles/blood , Indoles/chemistry , Indoles/pharmacology , Protein Kinase Inhibitors/chemistry , Syk Kinase
14.
Bioorg Med Chem Lett ; 21(16): 4745-9, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21764306

ABSTRACT

The structure-activity relationship of highly potent special ergolines which selectively block the chemokine receptor CXCR3 is reported. The most potent compounds showed IC(50) values below 10nM in both ligand binding and Ca(2+)-mobilization assays. However, these compounds were poorly active in an assay that measures receptor occupancy in blood. Introduction of polar substituents led to derivatives with IC(50) values below 10nM in this assay. Among them was compound 11a which showed both a favorable PK profile and cross reactivity with rodent CXCR3 making it a promising tool compound to further explore the role of CXCR3 in animal models.


Subject(s)
Ergolines/pharmacology , Receptors, CXCR3/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Ergolines/chemical synthesis , Ergolines/chemistry , Humans , Molecular Structure , Rats , Receptors, CXCR3/blood , Stereoisomerism , Structure-Activity Relationship
15.
Chem Biol ; 18(3): 314-23, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21439476

ABSTRACT

Genetic deficiency of Jak3 leads to abrogation of signal transduction through the common gamma chain (γc) and thus to immunodeficiency suggesting that specific inhibition of Jak3 kinase may result in immunosuppression. Jak1 cooperates with Jak3 in signaling through γc-containing receptors. Unexpectedly, a Jak3-selective inhibitor was less efficient in abolishing STAT5 phosphorylation than pan-Jak inhibitors. We therefore explored the roles of Jak1 and Jak3 kinase functionality in signaling using a reconstituted system. The presence of kinase-inactive Jak1 but not kinase-inactive Jak3 resulted in complete abolishment of STAT5 phosphorylation. Specific inhibition of the "analog-sensitive" mutant AS-Jak1 but not AS-Jak3 by the ATP-competitive analog 1NM-PP1 abrogated IL-2 signaling, corroborating the data with the selective Jak3 inhibitor. Jak1 thus plays a dominant role over Jak3 and these data challenge the notion that selective ATP-competitive Jak3 kinase inhibitors will be effective.


Subject(s)
Janus Kinase 1/metabolism , Janus Kinase 3/metabolism , Receptors, Cytokine/metabolism , Signal Transduction , Animals , Cell Line , Humans , Interleukin-2/pharmacology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/genetics , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/genetics , Mice , Mutation , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering , Receptors, Cytokine/chemistry , STAT5 Transcription Factor/metabolism
16.
J Med Chem ; 54(1): 284-8, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21155605

ABSTRACT

We describe a synthetic approach toward the rapid modification of phenyl-indolyl maleimides and the discovery of potent Jak3 inhibitor 1 with high selectivity within the Jak kinase family. We provide a rationale for this unprecedented selectivity based on the X-ray crystal structure of an analogue of 1 bound to the ATP-binding site of Jak3. While equally potent compared to the Pfizer pan Jak inhibitor CP-690,550 (2) in an enzymatic Jak3 assay, compound 1 was found to be 20-fold less potent in cellular assays measuring cytokine-triggered signaling through cytokine receptors containing the common γ chain (γC). Contrary to compound 1, compound 2 inhibited Jak1 in addition to Jak3. Permeability and cellular concentrations of compounds 1 and 2 were similar. As Jak3 always cooperates with Jak1 for signaling, we speculate that specific inhibition of Jak3 is not sufficient to efficiently block γC cytokine signal transduction required for strong immunosuppression.


Subject(s)
Indoles/chemical synthesis , Janus Kinase 3/antagonists & inhibitors , Maleimides/chemical synthesis , Cell Line , Cell Membrane Permeability , Crystallography, X-Ray , Humans , Immunosuppressive Agents/chemical synthesis , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Indoles/chemistry , Indoles/pharmacology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 3/chemistry , Maleimides/chemistry , Maleimides/pharmacology , Models, Molecular , Molecular Structure , Phosphorylation , Piperidines , Pyrimidines/pharmacology , Pyrroles/pharmacology , STAT5 Transcription Factor/metabolism , Structure-Activity Relationship
17.
J Immunol ; 186(1): 121-31, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21131428

ABSTRACT

Antagonism of CXCR4 disrupts the interaction between the CXCR4 receptor on hematopoietic stem cells (HSCs) and the CXCL12 expressed by stromal cells in the bone marrow, which subsequently results in the shedding of HSCs to the periphery. Because of their profound immunomodulatory effects, HSCs have emerged as a promising therapeutic strategy for autoimmune disorders. We sought to investigate the immunomodulatory role of mobilized autologous HSCs, via target of the CXCR4-CXL12 axis, to promote engraftment of islet cell transplantation. Islets from BALB/c mice were transplanted beneath the kidney capsule of hyperglycemic C57BL/6 mice, and treatment of recipients with CXCR4 antagonist resulted in mobilization of HSCs and in prolongation of islet graft survival. Addition of rapamycin to anti-CXCR4 therapy further promoted HSC mobilization and islet allograft survival, inducing a robust and transferable host hyporesponsiveness, while administration of an ACK2 (anti-CD117) mAb halted CXCR4 antagonist-mediated HSC release and restored allograft rejection. Mobilized HSCs were shown to express high levels of the negative costimulatory molecule programmed death ligand 1 (PD-L1), and HSCs extracted from wild-type mice, but not from PD-L1 knockout mice, suppressed the in vitro alloimmune response. Moreover, HSC mobilization in PD-L1 knockout mice failed to prolong islet allograft survival. Targeting the CXCR4-CXCL12 axis thus mobilizes autologous HSCs and promotes long-term survival of islet allografts via a PD-L1-mediated mechanism.


Subject(s)
B7-1 Antigen/physiology , Chemokine CXCL12/antagonists & inhibitors , Gene Targeting , Graft Survival/immunology , Hematopoietic Stem Cell Transplantation , Islets of Langerhans Transplantation/immunology , Membrane Glycoproteins/physiology , Peptides/physiology , Receptors, CXCR4/antagonists & inhibitors , Animals , B7-1 Antigen/genetics , B7-H1 Antigen , Benzylamines , Chemokine CXCL12/metabolism , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Cyclams , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/surgery , Gene Targeting/methods , Graft Survival/drug effects , Graft Survival/genetics , Heterocyclic Compounds/pharmacology , Islets of Langerhans Transplantation/pathology , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Peptides/deficiency , Peptides/genetics , Receptors, CXCR4/metabolism , Transplantation, Homologous
18.
Chimia (Aarau) ; 64(1-2): 23-8, 2010.
Article in English | MEDLINE | ID: mdl-21137679

ABSTRACT

Pig-to-human xenotransplantation of islet cells or of vascularized organs would offer a welcome treatment alternative for the ever-increasing number of patients with end-stage organ failure who are waiting for a suitable allograph. The main hurdle are preexisting antibodies, most of which are specific for 'Linear-B', carbohydrate epitopes terminated by the unbranched Gal-alpha(1,3)Gal disaccharide. These antibodies are responsible for the 'hyper-acute rejection' of the xenograft by complement mediated hemorrhage. For depletion of such antibodies we have developed an artificial injectable antigen, a glycopolymer (GAS914) with a charge neutral poly-lysine backbone (degree of polymerization n = 1000) and 25% of its side chains coupled to Linear-B-trisaccharide. With an average molecular weight of 400 to 500 kD, presenting 250 trisaccharide epitopes per molecule, this multivalent array binds anti-alphaGal antibodies with at least three orders of magnitude higher avidity on a per-saccharide basis than the monomeric epitope. In vivo experiments with non-human primates documented that rather low doses--1 to 5 mg/kg of GAS914 injected i.v.--efficiently reduce the load of anti-Linear-B antibodies quickly by at least 80%. This treatment can be repeated without any sensitization to GAS914. Interestingly, although the antibody levels start raising 12 h after injection, they do not reach pretreatment levels. The polymer is degraded and excreted within hours, with a minute fraction remaining in lymphoid tissue of anti-alphaGal producing animals only, probably binding to and inhibiting antibody-producing B-cells. The results of pig-to-non-human primate xenotransplantations established GAS914 as a relevant therapeutic option for pig-to-human transplantations as well. The synthesis of GAS914 was successfully scaled up to kg amounts needed for first clinical studies. Key was the use of galactosyl transferases and UDP-galactose for the synthesis of the trisaccharide.


Subject(s)
Antibodies, Heterophile/immunology , Antigen Presentation/immunology , Antigens, Heterophile/pharmacology , Disaccharides/immunology , Epitopes/immunology , Swine , Transplantation, Heterologous/immunology , Trisaccharides/pharmacology , Animals , Antigens, Heterophile/immunology , B-Lymphocytes/immunology , Carbohydrate Sequence , Graft Rejection/immunology , Graft Rejection/prevention & control , Humans , In Vitro Techniques , Islets of Langerhans Transplantation/immunology , Transplantation Immunology , Trisaccharides/immunology
19.
Bioorg Med Chem Lett ; 19(21): 6185-8, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19783143

ABSTRACT

The special ergoline 1 is a highly potent, selective antagonist of the chemokine receptor CXCR3. The surprising selectivity of this LSD-related compound can be explained by different electronic and steric properties of the ergoline core structure caused by the urea portion of the molecule. Discovery, biopharmaceutical properties and first derivatives of this promising lead compound are discussed.


Subject(s)
Ergolines/chemistry , Receptors, CXCR3/antagonists & inhibitors , Animals , Dogs , Drug Discovery , Ergolines/pharmacology , Humans , Mice , Microsomes, Liver/metabolism , Molecular Conformation , Rats , Receptors, CXCR3/metabolism , Structure-Activity Relationship
20.
J Med Chem ; 51(24): 7915-20, 2008 Dec 25.
Article in English | MEDLINE | ID: mdl-19053768

ABSTRACT

The interaction of the chemokine receptor CXCR4 with its ligand CXCL12 is involved in many biological processes such as hematopoesis, migration of immune cells, as well as in cancer metastasis. CXCR4 also mediates the infection of T-cells with X4-tropic HIV functioning as a coreceptor for the viral envelope protein gp120. Here, we describe highly potent, selective CXCR4 inhibitors that block CXCR4/CXCL12 interactions in vitro and in vivo as well as the infection of target cells by X4-tropic HIV.


Subject(s)
Receptors, CXCR4/chemistry , Thiourea/chemistry , Administration, Oral , Animals , Biological Availability , Chemistry, Pharmaceutical/methods , Chemokine CXCL12/chemistry , Chemokines/metabolism , Drug Design , HIV Envelope Protein gp120/chemistry , Humans , Inhibitory Concentration 50 , Models, Chemical , Rats , Receptors, CXCR4/antagonists & inhibitors , T-Lymphocytes/metabolism , T-Lymphocytes/virology
SELECTION OF CITATIONS
SEARCH DETAIL