72 results on '"Slack RJ"'
Search Results
2. Pharmacological characterization of GSK1004723, a novel, long-acting antagonist at histamine H1 and H3 receptors
- Author
-
Slack, RJ, Russell, LJ, Hall, DA, Luttmann, MA, Ford, AJ, Saunders, KA, Hodgson, ST, Connor, HE, Browning, C, and Clark, KL
- Subjects
Niacinamide ,Rhinitis, Allergic, Perennial ,Ovalbumin ,Bronchoconstriction ,Guinea Pigs ,Bronchi ,CHO Cells ,In Vitro Techniques ,Transfection ,Binding, Competitive ,Bronchial Provocation Tests ,Cell Line ,Cricetulus ,Piperidines ,Cricetinae ,Animals ,Humans ,Receptors, Histamine H3 ,Receptors, Histamine H1 ,Pyrilamine ,Allergens ,Benzazepines ,Research Papers ,Recombinant Proteins ,Histamine H1 Antagonists ,Phthalazines ,Carbachol ,Female ,Histamine ,Histamine H3 Antagonists - Abstract
Preclinical pharmacological characterization of GSK1004723, a novel, dual histamine H(1) and H(3) receptor antagonist.GSK1004723 was characterized in vitro and in vivo using methods that included radioligand binding, intracellular calcium mobilization, cAMP production, GTPγS binding, superfused human bronchus and guinea pig whole body plethysmography.In cell membranes over-expressing human recombinant H(1) and H(3) receptors, GSK1004723 displayed high affinity, competitive binding (H(1) pKi = 10.2; H(3) pKi = 10.6). In addition, GSK1004723 demonstrated slow dissociation from both receptors with a t(1/2) of 1.2 and 1.5 h for H(1) and H(3) respectively. GSK1004723 specifically antagonized H(1) receptor mediated increases in intracellular calcium and H(3) receptor mediated increases in GTPγS binding. The antagonism exerted was retained after cell washing, consistent with slow dissociation from H(1) and H(3) receptors. Duration of action was further evaluated using superfused human bronchus preparations. GSK1004723 (100 nmol·L(-1) ) reversed an established contractile response to histamine. When GSK1004723 was removed from the perfusate, only 20% recovery of the histamine response was observed over 10 h. Moreover, 21 h post-exposure to GSK1004723 there remained almost complete antagonism of responses to histamine. In vivo pharmacology was studied in conscious guinea pigs in which nasal congestion induced by intranasal histamine was measured indirectly (plethysmography). GSK1004723 (0.1 and 1 mg·mL(-1) intranasal) antagonized the histamine-induced response with a duration of up to 72 h.GSK1004723 is a potent and selective histamine H(1) and H(3) receptor antagonist with a long duration of action and represents a potential novel therapy for allergic rhinitis.
- Published
- 2011
3. Pharmacological characterization of GSK1004723, a novel, long-acting antagonist at histamine H1 and H3 receptors
- Author
-
Slack, RJ, primary, Russell, LJ, additional, Hall, DA, additional, Luttmann, MA, additional, Ford, AJ, additional, Saunders, KA, additional, Hodgson, ST, additional, Connor, HE, additional, Browning, C, additional, and Clark, KL, additional
- Published
- 2011
- Full Text
- View/download PDF
4. Pharmacological characterization of GB1908, a selective galectin-1 carbohydrate binding domain inhibitor for the treatment of cancer.
- Author
-
Herman KD, Holyer I, Humphries DC, Roper JA, Peterson K, Zetterberg FR, Pedersen A, Mackinnon AC, and Slack RJ
- Abstract
Introduction: Galectin-1 (Gal-1) is a lectin that has been shown to be involved in a number of pro-tumorigenic mechanisms and has also been shown to be immune-suppressive. Therefore, pharmacological blockade of Gal-1 has the potential to be therapeutically beneficial in cancers that overexpress this lectin where it is hypothesized to be driving cancer progression., Methods: GB1908 is a novel, selective and high affinity inhibitor of the Gal-1 carbohydrate recognition domain and in this study we have pharmacologically characterized this small molecule in a range of in vitro and in vivo systems in the context of cancer therapy. In addition, we used a data-driven approach to identify the cancer types which may benefit from Gal-1 inhibitor therapy., Results: The selectivity of GB1908 for Gal-1 compared with galectin-3 (Gal-3) was confirmed in biophysical and cellular assays. GB1908 attenuated Gal-1-induced T cell (Jurkat) apoptosis and reduced the production of immunosuppressive cytokines in a stromal non-small cell lung cancer (NSCLC) tumor microenvironment model. Breast carcinoma and metastatic skin cutaneous melanoma were identified as cancers in which high Gal-1 expression correlated with poorer survival outcomes in patients. Treatment with GB1908 slowed tumor growth in syngeneic mouse models of these cancers., Conclusion: The inhibition of both tumor growth and immune-suppressive cytokines, in cancers in which high Gal-1 is associated with poorer survival outcomes, suggests a potential therapeutic benefit for Gal-1 inhibitors such as GB1908., (S. Karger AG, Basel.)
- Published
- 2025
- Full Text
- View/download PDF
5. Development and Characterization of a High-Affinity Selective Galectin-3 Mouse Tool Compound in Mouse Models of Cancer.
- Author
-
Peterson K, Nilsson UJ, Gravelle L, Holyer I, Jansson K, Kahl-Knutson B, Leffler H, MacKinnon AC, Roper JA, Slack RJ, Wachenfeldt HV, Pedersen A, and Zetterberg FR
- Subjects
- Animals, Mice, Humans, Female, Antineoplastic Agents pharmacology, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents therapeutic use, Antineoplastic Agents chemical synthesis, Galectins antagonists & inhibitors, Galectins metabolism, Mice, Inbred C57BL, Cell Line, Tumor, Disease Models, Animal, Structure-Activity Relationship, Blood Proteins metabolism, Galectin 3 antagonists & inhibitors, Galectin 3 metabolism
- Abstract
The interest in galectin-3 as a drug target in the cancer and fibrosis space has grown during the past few years with several new classes of compounds being developed. The first orally available galectin-3 inhibitor, GB1211 (h-galectin-3 K
d = 0.025 μM), is currently in phase 2 clinical trials. Due to structural differences between human and mouse galectin-3 a significant reduction in mouse galectin-3 affinity is observed for most highly potent human galectin-3 inhibitors including GB1211 (m-galectin-3 Kd = 0.77 μM). Pharmacokinetic experiments in mouse dosing GB1211 up to 100 mg/kg results in free plasma levels below m-galectin-3 Kd , which is not comparable to the data observed in humans. To better support translation into clinical studies, a new improved mouse galectin-3 tool compound, GB2095 , was developed. Dosing this new compound in in vivo syngeneic mouse models of cancer resulted in reduction of the growth of breast and melanoma cancers.- Published
- 2024
- Full Text
- View/download PDF
6. Effect of GB1107, a novel galectin-3 inhibitor on pro-fibrotic signalling in the liver.
- Author
-
MacKinnon AC, Humphries DC, Herman K, Roper JA, Holyer I, Mabbitt J, Mills R, Nilsson UJ, Leffler H, Pedersen A, Schambye H, Zetterberg F, and Slack RJ
- Subjects
- Animals, Mice, Male, Mice, Inbred C57BL, Carbon Tetrachloride, Humans, Galectins metabolism, Hydrocarbons, Fluorinated, Triazoles, Galectin 3 metabolism, Galectin 3 antagonists & inhibitors, Galectin 3 genetics, Liver Cirrhosis drug therapy, Liver Cirrhosis pathology, Liver Cirrhosis metabolism, Liver Cirrhosis chemically induced, Signal Transduction drug effects, Liver drug effects, Liver metabolism, Liver pathology
- Abstract
Background and Purpose: Galectin-3 (Gal-3) is a pro-fibrotic β-galactoside binding lectin highly expressed in fibrotic liver and implicated in hepatic fibrosis. GB1107 is a novel orally active Gal-3 small molecule inhibitor that has high affinity for Gal-3 >1000-fold selectively over other galectins. The aim of this study was to characterise GB1107 and galectin-3 in vitro and in vivo in the context of fibrosis signalling and liver disease., Experimental Approach: Liver fibrosis was induced by administration of CCl
4 twice weekly by intraperitoneal injection in mice for 8 weeks. GB1107 was orally administered once daily (10 mg/kg) for the last 4 weeks of CCl4 treatment. Fibrosis was assessed by picrosirius red staining of FFPE sections. Liver enzymes, Gal-3 and downstream biomarkers were assessed in liver and plasma. Paired-end sequencing was performed on the Nextseq 2000 platform. Pathway enrichment analysis was performed to determine enrichment of differentially expressed genes (DEGs) within Reactome pathways and Gene Ontology (GO) terms., Key Results: GB1107 significantly reduced plasma transaminases and liver Gal-3 and reduced liver fibrosis. RNAseq analysis of whole liver showed that 1659 DEGs were identified with CCl4 treatment compared to control. Pathways enriched in up-regulated genes in the CCl4 group included those related to the extracellular matrix, collagen biosynthesis and assembly, cell cycle and the immune system. Comparing GB1107 treatment with CCl4 control 1147 DEGs were identified. GB1107 effectively reversed the majority of the CCl4 induced gene changes., Conclusions and Implications: GB1107 attenuated liver fibrosis and highlights Gal-3 as a therapeutic target for hepatic fibrosis., Competing Interests: Declaration of competing interest Conflict of interest disclosure: DH, JR, IH, JM, RJS, ACM, AP, FZ, HS: employees, personal fees – Galecto Biotech AB, UJN, HL: shareholders, personal fees – Galecto Biotech AB., (Copyright © 2024 Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
7. Core Modifications of GSK3335103 toward Orally Bioavailable α v β 6 Inhibitors with Improved Synthetic Tractability.
- Author
-
Hryczanek HF, Barrett J, Barrett TN, Burley GA, Cookson RE, Hatley RJD, Measom ND, Roper JA, Rowedder JE, Slack RJ, Śmieja CB, and Macdonald SJF
- Subjects
- Administration, Oral, Animals, Humans, Structure-Activity Relationship, Rats, Male, Mice, Antigens, Neoplasm, Biological Availability, Integrins antagonists & inhibitors, Integrins metabolism
- Abstract
The α
v β6 integrin has been identified as a target for the treatment of fibrotic diseases, based on the role it has in activating TGF-β1 , a protein implicated in the pathogenesis of fibrosis. However, the development of orally bioavailable αv β6 inhibitors has proven challenging due to the zwitterionic pharmacophore required to bind to the RGD binding site. This work describes the design and development of a novel, orally bioavailable series of αv β6 inhibitors, developing on two previously published αv β6 inhibitors, GSK3008348 and GSK3335103. Strategies to reduce the basicity of the central ring nitrogen present in GSK3008348 were employed, while avoiding the synthetic complexity of the chiral, fluorine-containing quaternary carbon center contained in GSK3335103. Following initial PK studies, this series was optimized, aided by analysis of the physicochemical and in vitro PK properties, to deliver lead molecules ( S )- 20 and 28 as potent and orally bioavailable αv β6 inhibitors with improved synthetic tractability.- Published
- 2024
- Full Text
- View/download PDF
8. Galectin-1 Induces the Production of Immune-Suppressive Cytokines in Human and Mouse T Cells.
- Author
-
Herman KD, Holyer I, Humphries DC, Adamska A, Roper JA, Peterson K, Zetterberg FR, Pedersen A, MacKinnon AC, and Slack RJ
- Subjects
- Animals, Humans, Mice, Concanavalin A pharmacology, Mice, Inbred C57BL, Cells, Cultured, Galectin 1 metabolism, Galectin 1 genetics, Cytokines metabolism, T-Lymphocytes immunology, T-Lymphocytes metabolism, T-Lymphocytes drug effects
- Abstract
Galectin-1 is implicated in several pro-tumourigenic mechanisms and is considered immune-suppressive. The pharmacological inhibition of galectin-1 may be beneficial in cancers in which galectin-1 is overexpressed and driving cancer progression. This study aimed to further characterise the immunosuppressive cytokines influenced by galectin-1 in in vitro immune cell cultures and an in vivo inflammatory model using a recently discovered selective inhibitor of galectin-1, GB1908. To enable a translational approach and link mouse and human pharmacology, anti-CD3/anti-CD28 stimulated T cells cultured from human whole blood and mouse spleens were compared. For in vivo studies of T cell-mediated inflammation, the concanavalin-A (Con-A) mouse model was used to induce a T lymphocyte-driven acute liver injury phenotype. The inhibition of galectin-1 with GB1908 reduced IL-17A, IFNγ and TNFα in a concentration-dependent manner in both mouse and human T cells in vitro. The immunosuppressive cytokines measured in Con-A-treated mice were all upregulated compared to naïve mice. Subsequently, mice treated with GB1908 demonstrated a significant reduction in IL-17A, IFNγ, IL-6 and TNFα compared to vehicle-treated mice. In conclusion, galectin-1 induced the production of several important immune-suppressive cytokines from T cells in vitro and in vivo. This result suggests that, in the context of cancer therapy, a selective galectin-1 could be a viable approach as a monotherapy, or in combination with chemotherapeutic agents and/or checkpoint inhibitors, to enhance the numbers and activity of cytotoxic T cells in the tumour microenvironment of high galectin-1 expressing cancers.
- Published
- 2024
- Full Text
- View/download PDF
9. Relative bioavailability and food effect of the galectin-3 inhibitor selvigaltin (GB1211) administered as a tablet in healthy participants (GALBA-1).
- Author
-
Aslanis V, Abd-Elaziz K, Slack RJ, Brinch A, Gravelle L, Morley W, Phung, Herman K, Holyer I, Poulsen KK, Dogterom P, Tantawi S, Zetterberg FR, Jacoby B, Schambye H, and Lindmark B
- Subjects
- Humans, Male, Adult, Female, Young Adult, Middle Aged, Administration, Oral, Capsules, Fasting, Galectin 3 antagonists & inhibitors, Area Under Curve, Blood Proteins metabolism, Galectins antagonists & inhibitors, Cross-Over Studies, Biological Availability, Tablets, Food-Drug Interactions, Healthy Volunteers
- Abstract
Purpose: Overexpression of galectin-3, a β-galactoside-binding lectin, is associated with fibrotic diseases and cancer. Selvigaltin is an oral galectin-3 inhibitor, previously administered as a 50 mg capsule. This study aimed to evaluate the relative bioavailability and food effect of selvigaltin as a 100 mg tablet in healthy volunteers., Methods: In this single-dose, randomized, three-period, crossover study (GALBA-1; NCT05747573), participants received selvigaltin as a 100 mg tablet (under fasted and fed conditions) or as two 50 mg capsules (under fasted conditions). Primary endpoints included plasma and urine pharmacokinetic (PK) parameters. Secondary endpoints were safety and tolerability., Results: Of the 13 enrolled participants, 12 completed the study. Under fasted conditions, geometric mean maximum observed plasma concentration (C
max ) and systemic exposure (AUC0─inf ) of selvigaltin were 161.0% and 84.0% higher, respectively, after administration of a tablet vs. capsules. Under fed vs. fasted conditions, geometric mean Cmax of the selvigaltin tablet was 20.0% lower, whereas AUC0─inf was unaffected. Geometric mean percentage of total dose of selvigaltin excreted in urine over 0─96 h was 30.3% and 35.9% for the tablet under fasted and fed conditions, respectively, and 14.5% for the capsules. No treatment-emergent severe or serious adverse events or study discontinuations due to a treatment-emergent adverse event were reported., Conclusion: The tablet formulation of selvigaltin displayed higher bioavailability vs. the capsule formulation, with minimal effect of food on PK. Selvigaltin was well-tolerated during all treatments. These findings warrant further clinical development of the tablet formulation of selvigaltin without specific food restrictions., Clinical Trial Registration: NCT05747573; February 28, 2023., (© 2024. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)- Published
- 2024
- Full Text
- View/download PDF
10. Discovery and Development of Highly Potent and Orally Bioavailable Nonpeptidic α v β 6 Integrin Inhibitors.
- Author
-
Procopiou PA, Barrett J, Crawford MHJ, Hatley RJD, Hancock AP, Pritchard JM, Rowedder JE, Copley RCB, Slack RJ, Sollis SL, Thorp LR, Lippa RA, Macdonald SJF, and Barrett TN
- Subjects
- Animals, Dogs, Rats, Administration, Oral, Humans, Swine, Structure-Activity Relationship, Drug Discovery, Swine, Miniature, Male, Rats, Sprague-Dawley, Integrins antagonists & inhibitors, Integrins metabolism, Biological Availability, Antigens, Neoplasm metabolism
- Abstract
A series of 3-aryl(( S )-3-fluoropyrrolidin-1-yl)butanoic acids were developed as potent orally bioavailable α
v β6 integrin inhibitors. Starting from a zwitterionic peptidomimetic series optimized for inhaled administration, the balancing of potency and passive permeability to achieve suitable oral agents through modification and exploration of aryl substituents and p Ka of the central cyclic amine is described. ( S )-4-(( S )-3-Fluoro-3-(2-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)ethyl)pyrrolidin-1-yl)-3-(3-(2-methoxyethoxy)phenyl)butanoic acid was found to have highly desirable oral pharmacokinetic profiles in rat, dog, and minipig, with low to moderate clearance (26%, 7%, and 18% liver blood flow, respectively), moderate volumes of distribution (3.6, 1.4, and 0.9 L/kg, respectively), high to complete oral bioavailabilities, high αv β6 integrin potency of pIC50 of 8.0, and high solubility in physiological media (>2 mg/mL). Equating to the estimated human dose range of 10-75 mg b.i.d. to achieve 90% αv β6 target engagement at Cmin , it was selected for further investigation as a potential therapeutic agent for the treatment of idiopathic pulmonary fibrosis.- Published
- 2024
- Full Text
- View/download PDF
11. Single‑Dose Pharmacokinetics and Safety of the Oral Galectin‑3 Inhibitor, Selvigaltin (GB1211), in Participants with Hepatic Impairment.
- Author
-
Aslanis V, Gray M, Slack RJ, Zetterberg FR, Tonev D, Phung, Smith B, Jacoby B, Schambye H, Krastev Z, Ungell AL, and Lindmark B
- Subjects
- Humans, Male, Female, Middle Aged, Administration, Oral, Aged, Adult, Blood Proteins metabolism, Liver Diseases metabolism, Galectins antagonists & inhibitors, Galectin 3 antagonists & inhibitors, Galectin 3 blood
- Abstract
Background and Objectives: Selvigaltin (GB1211), an orally available small molecule galectin-3 inhibitor developed as a treatment for liver fibrosis and cirrhosis, was evaluated to assess the effect of hepatic impairment on its pharmacokinetics and safety to address regulatory requirements., Methods: GULLIVER-2 was a Phase Ib/IIa three-part study. Parts 1 and 3 had single-dose, open-label designs assessing pharmacokinetics (plasma [total and unbound] and urine), safety, and tolerability of 100 mg oral selvigaltin in participants with moderate (Child-Pugh B, Part 1) or severe (Child-Pugh C, Part 3) hepatic impairment, compared with healthy-matched participants (n = 6 each)., Results: All participants received selvigaltin and completed the study. No adverse events were reported. The median time to reach maximum total plasma concentration following drug administration was of 3.49 and 4.00 h post-dose for Child-Pugh B and C participants, respectively; comparable with controls. Total plasma exposure was higher for participants with hepatic impairment compared with controls. Whilst maximum plasma concentration (C
max ) was unaffected in Child-Pugh B participants, area under the plasma concentration-time curve from time zero to infinity (AUC∞ ) increased by ~ 1.7-fold compared with controls, and half-life was prolonged (geometric mean 28.15 vs 16.38 h). In Child-Pugh C participants, Cmax increased by ~ 1.3-fold, AUC∞ increased by ~ 1.5-fold, and half-life was prolonged (21.05 vs 16.14 h). No trend was observed in plasma unbound fractions or urinary excretion of unchanged selvigaltin in either group., Conclusion: Hepatic impairment increased selvigaltin exposure without safety concerns. These data can inform dose recommendations for future clinical programmes., Trial Registration: Clinicaltrials.gov NCT05009680., (© 2024. The Author(s), under exclusive licence to Springer Nature Switzerland AG.)- Published
- 2024
- Full Text
- View/download PDF
12. The galectin-3 inhibitor selvigaltin reduces liver inflammation and fibrosis in a high fat diet rabbit model of metabolic-associated steatohepatitis.
- Author
-
Comeglio P, Guarnieri G, Filippi S, Cellai I, Acciai G, Holyer I, Zetterberg F, Leffler H, Kahl-Knutson B, Sarchielli E, Morelli A, Maggi M, Slack RJ, and Vignozzi L
- Abstract
Introduction: Galectin-3 is a pro-fibrotic β-galactoside binding lectin highly expressed in fibrotic liver and implicated in hepatic fibrosis. Selvigaltin (previously known as GB1211) is a novel orally active galectin-3 small molecule inhibitor that has high affinity for galectin-3 (human K
D = 25 nM; rabbit KD = 12 nM) and high oral bioavailability in rabbits and man. In this study the efficacy of selvigaltin was investigated in a high fat diet (HFD) rabbit model of metabolic-associated steatohepatitis (MASH)., Methods: Male New Zealand White rabbits were individually caged under standard conditions in a temperature and humidity-controlled room on a 12 h light/darkness cycle. After 1 week of regular diet (RD), rabbits were randomly assigned for 8 or 12 weeks to different groups: RD/vehicle, RD/selvigaltin, HFD (8 weeks), HFD/vehicle and HFD/selvigaltin (0.3, 1.0, 5.0 or 30 mg/kg selvigaltin with vehicle/selvigaltin p.o. dosed therapeutically q.d. 5 days per week from week 9 or 12). Liver inflammation, steatosis, ballooning, and fibrosis was measured via blood metabolic markers, histomorphological evaluation [Oil Red O, Giemsa, Masson's trichome, picrosirius red (PSR) and second harmonic generation (SHG)], and mRNA and protein expression., Results: Steatosis, inflammation, ballooning, and fibrosis were all increased from RD to HFD/vehicle groups. Selvigaltin demonstrated target engagement by significantly decreasing galectin-3 levels in the liver as measured via immunohistochemistry and mRNA analysis. Selvigaltin dose-dependently reduced biomarkers of liver function (AST, ALT, bilirubin), inflammation (cells foci), and fibrosis (PSR, SHG), as well as decreasing the mRNA and protein expression of several key inflammation and fibrosis biomarkers (e.g., IL6, TGFβ3, SNAI2, collagen). Doses of 1.0 or 5.0 mg/kg demonstrated consistent efficacy across most biological endpoints supporting the current clinical doses of selvigaltin being investigated in liver disease., Discussion: Selvigaltin significantly reduced hepatic inflammation and fibrosis in an HFD rabbit model of MASH following therapeutic dosing for 4 weeks in a dose-dependent manner. These data support the human selvigaltin dose of 100 mg b.i.d. that has been shown to reduce key liver biomarkers during a clinical study in liver cirrhosis., Competing Interests: Author(s) IH, FZ, HL, and RS were employed by Galecto Biotech AB. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The authors declare that this study received funding from Galecto Biotech AB. The funder was involved in the study design, interpretation of data, the writing of this article, and the decision to submit it for publication., (Copyright © 2024 Comeglio, Guarnieri, Filippi, Cellai, Acciai, Holyer, Zetterberg, Leffler, Kahl-Knutson, Sarchielli, Morelli, Maggi, Slack and Vignozzi.)- Published
- 2024
- Full Text
- View/download PDF
13. A Quantitative Human Red Blood Cell Agglutination Assay for Characterisation of Galectin Inhibitors.
- Author
-
Gasson R, Roper JA, and Slack RJ
- Subjects
- Humans, Galectin 3 antagonists & inhibitors, Galectin 3 metabolism, Agglutination Tests methods, Hemagglutination Tests, Agglutination drug effects, Erythrocytes metabolism, Erythrocytes drug effects, Hemagglutination drug effects, Galectins antagonists & inhibitors, Galectins metabolism, Galectin 1 antagonists & inhibitors, Galectin 1 metabolism
- Abstract
Galectins are a family of beta-galactoside-binding proteins that are characterised by their carbohydrate recognition domain (CRD) and include galectin-1 and galectin-3. These galectins have been implicated in numerous diseases due to their pleiotropic nature, including cancer and fibrosis, with therapeutic inhibitors being clinically developed to block the CRD. One of the early methods developed to characterise these galectins was the hemagglutination of red blood cells. Although it is insightful, this approach has been hampered by a lack of sensitivity and accurate quantification of the agglutination observed. In this study, we aimed to validate a more precise and quantitative method to enable the further investigation of differences between galectins in respect to agglutination induction in different blood groups, as well as the characterisation of small molecule inhibitors. Quantification of hemagglutination was shown to be optimal using U-bottom plates imaged and analysed with FIJI ImageJ rather than flat-bottom plates read for absorbance on an optical density plate reader. Galectin-3-induced red blood cell agglutination efficacy increased significantly from blood group O to A to B. However, for both the galectin-1 monomer and concatemer, a more comparable effect was observed between blood group B and O, but with more potent effects than in blood group A. Inhibition assays for both galectin-3 and galectin-1 induced-hemagglutination were able to demonstrate clear concentration responses and expected selectivity profiles for a set of small-molecule glycomimetics, confirming the historical profiles obtained in biochemical binding and functional cellular assays.
- Published
- 2024
- Full Text
- View/download PDF
14. Determining the Affinity and Kinetics of Small Molecule Inhibitors of Galectin-1 Using Surface Plasmon Resonance.
- Author
-
Kim H, Kretz L, Ronin C, Starck C, Roper JA, Kahl-Knutson B, Peterson K, Leffler H, Nilsson UJ, Pedersen A, Zetterberg FR, and Slack RJ
- Subjects
- Humans, Animals, Mice, Kinetics, Small Molecule Libraries pharmacology, Small Molecule Libraries chemistry, Fluorescence Polarization methods, Galectin 1 metabolism, Galectin 1 antagonists & inhibitors, Galectin 1 chemistry, Surface Plasmon Resonance methods, Protein Binding
- Abstract
The beta-galactoside-binding mammalian lectin galectin-1 can bind, via its carbohydrate recognition domain (CRD), to various cell surface glycoproteins and has been implicated in a range of cancers. As a consequence of binding to sugar residues on cell surface receptors, it has been shown to have a pleiotropic effect across many cell types and mechanisms, resulting in immune system modulation and cancer progression. As a result, it has started to become a therapeutic target for both small and large molecules. In previous studies, we used fluorescence polarization (FP) assays to determine K
D values to screen and triage small molecule glycomimetics that bind to the galectin-1 CRD. In this study, surface plasmon resonance (SPR) was used to compare human and mouse galectin-1 affinity measures with FP, as SPR has not been applied for compound screening against this galectin. Binding affinities for a selection of mono- and di-saccharides covering a 1000-fold range correlated well between FP and SPR assay formats for both human and mouse galectin-1. It was shown that slower dissociation drove the increased affinity at human galectin-1, whilst faster association was responsible for the effects in mouse galectin-1. This study demonstrates that SPR is a sound alternative to FP for early drug discovery screening and determining affinity estimates. Consequently, it also allows association and dissociation constants to be measured in a high-throughput manner for small molecule galectin-1 inhibitors.- Published
- 2024
- Full Text
- View/download PDF
15. Discovery of the Selective and Orally Available Galectin-1 Inhibitor GB1908 as a Potential Treatment for Lung Cancer.
- Author
-
Zetterberg FR, Peterson K, Nilsson UJ, Andréasson Dahlgren K, Diehl C, Holyer I, Håkansson M, Khabut A, Kahl-Knutson B, Leffler H, MacKinnon AC, Roper JA, Slack RJ, Zarrizi R, and Pedersen A
- Subjects
- Humans, Animals, Mice, Administration, Oral, Apoptosis drug effects, Structure-Activity Relationship, Jurkat Cells, Drug Discovery, Crystallography, X-Ray, Thiazoles pharmacokinetics, Thiazoles pharmacology, Thiazoles chemistry, Galectin 1 antagonists & inhibitors, Galectin 1 metabolism, Lung Neoplasms drug therapy, Lung Neoplasms pathology, Antineoplastic Agents pharmacology, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents chemistry, Antineoplastic Agents therapeutic use
- Abstract
We have previously described a new series of selective and orally available galectin-1 inhibitors resulting in the thiazole-containing glycomimetic GB1490. Here, we show that the introduction of polar substituents to the thiazole ring results in galectin-1-specific compounds with low nM affinities. X-ray structural analysis of a new ligand-galectin-1 complex shows changes in the binding mode and ligand-protein hydrogen bond interactions compared to the GB1490-galectin-1 complex. These new high affinity ligands were further optimized with respect to affinity and ADME properties resulting in the galectin-1-selective GB1908 ( K
d galectin-1/3 0.057/6.0 μM). In vitro GB1908 inhibited galectin-1-induced apoptosis in Jurkat cells (IC50 = 850 nM). Pharmacokinetic experiments in mice revealed that a dose of 30 mg/kg b.i.d. results in free levels of GB1908 in plasma over galectin-1 Kd for 24 h. GB1908 dosed with this regimen reduced the growth of primary lung tumor LL/2 in a syngeneic mouse model.- Published
- 2024
- Full Text
- View/download PDF
16. Defining the mechanism of galectin-3-mediated TGF-β1 activation and its role in lung fibrosis.
- Author
-
Calver JF, Parmar NR, Harris G, Lithgo RM, Stylianou P, Zetterberg FR, Gooptu B, Mackinnon AC, Carr SB, Borthwick LA, Scott DJ, Stewart ID, Slack RJ, Jenkins RG, and John AE
- Subjects
- Humans, Lung metabolism, Lung pathology, Signal Transduction, Receptor, Transforming Growth Factor-beta Type II metabolism, Receptor, Transforming Growth Factor-beta Type II genetics, Receptors, Transforming Growth Factor beta metabolism, Protein Binding, Protein Serine-Threonine Kinases metabolism, Galectins metabolism, Collagen Type I metabolism, Cells, Cultured, Blood Proteins, Transforming Growth Factor beta1 metabolism, Galectin 3 metabolism, Galectin 3 genetics, Fibroblasts metabolism, Fibroblasts pathology, Idiopathic Pulmonary Fibrosis metabolism, Idiopathic Pulmonary Fibrosis pathology
- Abstract
Integrin-mediated activation of the profibrotic mediator transforming growth factor-β1 (TGF-β1), plays a critical role in idiopathic pulmonary fibrosis (IPF) pathogenesis. Galectin-3 is believed to contribute to the pathological wound healing seen in IPF, although its mechanism of action is not precisely defined. We hypothesized that galectin-3 potentiates TGF-β1 activation and/or signaling in the lung to promote fibrogenesis. We show that galectin-3 induces TGF-β1 activation in human lung fibroblasts (HLFs) and specifically that extracellular galectin-3 promotes oleoyl-L-α-lysophosphatidic acid sodium salt-induced integrin-mediated TGF-β1 activation. Surface plasmon resonance analysis confirmed that galectin-3 binds to αv integrins, αvβ1, αvβ5, and αvβ6, and to the TGFβRII subunit in a glycosylation-dependent manner. This binding is heterogeneous and not a 1:1 binding stoichiometry. Binding interactions were blocked by small molecule inhibitors of galectin-3, which target the carbohydrate recognition domain. Galectin-3 binding to β1 integrin was validated in vitro by coimmunoprecipitation in HLFs. Proximity ligation assays indicated that galectin-3 and β1 integrin colocalize closely (≤40 nm) on the cell surface and that colocalization is increased by TGF-β1 treatment and blocked by galectin-3 inhibitors. In the absence of TGF-β1 stimulation, colocalization was detectable only in HLFs from IPF patients, suggesting the proteins are inherently more closely associated in the disease state. Galectin-3 inhibitor treatment of precision cut lung slices from IPF patients' reduced Col1a1, TIMP1, and hyaluronan secretion to a similar degree as TGF-β type I receptor inhibitor. These data suggest that galectin-3 promotes TGF-β1 signaling and may induce fibrogenesis by interacting directly with components of the TGF-β1 signaling cascade., Competing Interests: Conflict of interests R. G. J. is a trustee for Action for Pulmonary Fibrosis. A. E. J. is a founder and shareholder of Alevin Therapeutics. J. F. C., F. R. Z., A. C. M., and R. J. S. are Galecto employees with shares/options in the company. N. R. P. is a Roche employee. L. A. B. is a director and shareholder of FibroFind. G. H., R. M. L., P. S., S. B. C., D. J. S., and I. D. S. declare that they have no conflict of interests with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
17. Structured Weaning From the Impella Left Ventricular Micro-Axial Pump in Acute Myocardial Infarction With Cardiogenic Shock and Protected Percutaneous Coronary Intervention: Experience From a Non-Cardiac Surgical Centre.
- Author
-
Slack RJ, McGain F, Cox N, French C, Cheng V, Stub D, Zakhem B, Dade F, Bloom JE, Chan W, and Yang Y
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Ventricular Function, Left physiology, Retrospective Studies, Echocardiography, Follow-Up Studies, Shock, Cardiogenic therapy, Shock, Cardiogenic surgery, Heart-Assist Devices, Percutaneous Coronary Intervention methods, Myocardial Infarction complications
- Abstract
Background: The Impella (Abiomed, Danvers, MA, USA) temporary percutaneous left ventricular assist device is increasingly used as mechanical circulatory support in patients with acute myocardial infarction-cardiogenic shock (AMICS) or those undergoing high-risk protected percutaneous coronary intervention (PCI). The optimal weaning regimen remains to be defined., Method: We implemented a structured weaning protocol in a series of 10 consecutive patients receiving Impella support for protected PCI or AMICS treated with PCI in a high volume non-cardiac surgery centre. Weaning after revascularisation was titrated to native heart recovery using both haemodynamic and echocardiographic parameters., Results: Ten patients (eight male, two female; aged 43-70 years) received Impella support for AMICS (80%) or protected PCI (20%). Cardiogenic shock was of Society for Cardiac Angiography & Interventions grade C-E of severity in 80%, and median left ventricular end-diastolic pressure was 31 mmHg. Protocol implementation allowed successful weaning in eight of 10 patients with a median support time of 29 hours (range, 4-48 hours). Explantation was associated with an increase in heart rate (81 vs 88 bpm; p=0.005), but no significant change in Cardiac Index (2.9 vs 2.9 L/min/m
2 ), mean arterial pressure (79 vs 82 mmHg), vasopressor requirement (10% vs 10%), or serum lactate (1.0 vs 1.0). Median durations of intensive care and hospital stay were 3 and 6 days, respectively. At 30 days, the mortality rate was 20%, with median left ventricular ejection fraction of 40%., Conclusions: A structured and dynamic weaning protocol for patients with AMICS and protected PCI supported by the Impella device is feasible in a non-cardiac surgery centre. Larger studies are needed to assess generalisability of such a weaning protocol., Competing Interests: Conflicts of Interest There are no conflicts of interest to disclose., (Copyright © 2023 Australian and New Zealand Society of Cardiac and Thoracic Surgeons (ANZSCTS) and the Cardiac Society of Australia and New Zealand (CSANZ). Published by Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
18. Discovery of Selective and Orally Available Galectin-1 Inhibitors.
- Author
-
Zetterberg FR, Diehl C, Håkansson M, Kahl-Knutson B, Leffler H, Nilsson UJ, Peterson K, Roper JA, and Slack RJ
- Subjects
- Animals, Humans, Mice, Binding Sites, Carbohydrates chemistry, Jurkat Cells, Galectin 1 antagonists & inhibitors, Galectin 3 metabolism, Hydrocarbons, Fluorinated chemistry, Hydrocarbons, Fluorinated pharmacokinetics, Hydrocarbons, Fluorinated pharmacology, Triazoles chemistry, Triazoles pharmacokinetics, Triazoles pharmacology
- Abstract
A new series of orally available α-d-galactopyranosides with high affinity and specificity toward galectin-1 have been discovered. High affinity and specificity were achieved by changing six-membered aryl-triazolyl substituents in a series of recently published galectin-3-selective α-d-thiogalactosides (e.g., GB1107 K
d galectin-1/3 3.7/0.037 μM) for five-membered heterocycles such as thiazoles. The in vitro pharmacokinetic properties were optimized, resulting in several galectin-1 inhibitors with favorable properties. One compound, GB1490 ( Kd galectin-1/3 0.4/2.7 μM), was selected for further characterization toward a panel of galectins showing a selectivity of 6- to 320-fold dependent on galectin. The X-ray structure of GB1490 bound to galectin-1 reveals the compound bound in a single conformation in the carbohydrate binding site. GB1490 was shown to reverse galectin-1-induced apoptosis of Jurkat cells at low μM concentrations. No cell cytotoxicity was observed for GB1490 up to 90 μM in the A549 cells. In pharmacokinetic studies in mice, GB1490 showed high oral bioavailability ( F % > 99%).- Published
- 2023
- Full Text
- View/download PDF
19. Violence in intensive care: a point prevalence study.
- Author
-
Slack RJ, French C, McGain F, Bates S, Gao A, Knowles S, and Yang Y
- Abstract
Introduction: Violence in the intensive care unit (ICU) is poorly characterised and its incidence is largely extrapolated from studies in the emergency department. Policy requirements vary between jurisdictions and have not been formally evaluated. Methods: A multisite, single-time point observational study was conducted across Australasian ICUs which focused on the incidence of violence in the previous 24 hours, the characteristics of patients displaying violent behaviour, the perceived contributors, and the management strategies implemented. Unit policies were surveyed across a range of domains relevant to violence management. Results: Data were available for 627 patients admitted to 44 ICUs on one of 2 days in June 2019. Four per cent (25/627) displayed at least one episode of violent behaviour in the previous 24 hours. Violent behaviour was more likely in individuals after a greater length of stay in hospital (incidence, 2%, 4% and 7% for day 0-2, 3-7 and > 7 days respectively; P = 0.01) and in the ICU (2%, 4% and 9% for day 0-2, 3-7 and > 7 of ICU stay respectively; P < 0.01). The most common perceived contributors to violence were confusion (64%), physical illness (40%), and psychiatric illness (34%). Management with chemical sedation (72%) and physical restraint (28%) was commonly required. Clinicians assessed an additional 53 patients (53/627, 9%) as at risk of displaying violence in the next 24 hours. Of the 44 participating ICUs, 30 (68%) had a documented violence procedure. Conclusion: Violence in the ICU was common and frequently required intervention. In this study, one-third of ICUs did not have formal violence procedures, and in those with violence procedures, considerable variation was observed., Competing Interests: All authors declare that they do not have any potential conflict of interest in relation to this manuscript., (© 2022 College of Intensive Care Medicine of Australia and New Zealand.)
- Published
- 2023
- Full Text
- View/download PDF
20. Resistance to anti-PD-1/anti-PD-L1: galectin-3 inhibition with GB1211 reverses galectin-3-induced blockade of pembrolizumab and atezolizumab binding to PD-1/PD-L1.
- Author
-
Mabbitt J, Holyer ID, Roper JA, Nilsson UJ, Zetterberg FR, Vuong L, Mackinnon AC, Pedersen A, and Slack RJ
- Subjects
- Animals, Mice, Antibodies, Blocking, Galectin 3, Antibodies, Monoclonal, Humanized pharmacology, Antibodies, Monoclonal, Humanized therapeutic use
- Abstract
Background: Galectin-3 (Gal-3) is a β-galactoside-binding lectin that is highly expressed within the tumor microenvironment of aggressive cancers and has been suggested to predict a poor response to immune checkpoint therapy with the anti-PD-1 monoclonal antibody pembrolizumab. We aimed to assess if the effect of Gal-3 was a result of direct interaction with the immune checkpoint receptor., Methods: The ability of Gal-3 to interact with the PD-1/PD-L1 complex in the absence and presence of blocking antibodies was assessed in in vitro biochemical and cellular assays as well as in an in vivo syngeneic mouse cancer model., Results: Gal-3 reduced the binding of the checkpoint inhibitors pembrolizumab (anti-PD-1) and atezolizumab (anti-PD-L1), by potentiating the interaction between the PD-1/PD-L1 complex. In the presence of a highly selective Gal-3 small molecule inhibitor (GB1211) the binding of the anti-PD-1/anti-PD-L1 therapeutics was restored to control levels. This was observed in both a surface plasmon resonance assay measuring protein-protein interactions and via flow cytometry. Combination therapy with GB1211 and an anti-PD-L1 blocking antibody reduced tumor growth in an in vivo syngeneic model and increased the percentage of tumor infiltrating T lymphocytes., Conclusion: Our study suggests that Gal-3 can potentiate the PD-1/PD-L1 immune axis and potentially contribute to the immunosuppressive signalling mechanisms within the tumor microenvironment. In addition, Gal-3 prevents atezolizumab and pembrolizumab target engagement with their respective immune checkpoint receptors. Reversal of this effect with the clinical candidate GB1211 offers a potential enhancing combination therapeutic with anti-PD-1 and -PD-L1 blocking antibodies., Competing Interests: UN, FZ, RS, JR, IH, and AM have ownership interest including stock, patents, etc. in Galecto Biotech AB. AP is the COO at Galecto Biotech and has ownership interest including stock, patents, etc. in Galecto Biotech. The remaining author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Mabbitt, Holyer, Roper, Nilsson, Zetterberg, Vuong, Mackinnon, Pedersen and Slack.)
- Published
- 2023
- Full Text
- View/download PDF
21. Evaluating the affinity and kinetics of small molecule glycomimetics for human and mouse galectin-3 using surface plasmon resonance.
- Author
-
Kim H, Weidner N, Ronin C, Klein E, Roper JA, Kahl-Knutson B, Peterson K, Leffler H, Nilsson UJ, Pedersen A, Zetterberg FR, and Slack RJ
- Subjects
- Humans, Animals, Mice, Kinetics, Galectins chemistry, Galectins metabolism, Carbohydrates chemistry, Mammals metabolism, Galectin 3 genetics, Galectin 3 chemistry, Galectin 3 metabolism, Surface Plasmon Resonance
- Abstract
Galectin-3 is a beta-galactoside-binding mammalian lectin that is one of a 15-member galectin family that can bind several cell surface glycoproteins via its carbohydrate recognition domain (CRD). As a result, it can influence a range of cellular processes including cell activation, adhesion and apoptosis. Galectin-3 has been implicated in various diseases, including fibrotic disorders and cancer, and is now being therapeutically targeted by both small and large molecules. Historically, the screening and triaging of small molecule glycomimetics that bind to the galectin-3 CRD has been completed in fluorescence polarisation (FP) assays to determine K
D values. Surface plasmon resonance (SPR) has not been widely used for compound screening and in this study it was used to compare human and mouse galectin-3 affinity measures between FP and SPR, as well as investigate compound kinetics. The KD estimates for a set of compounds selected from mono- and di-saccharides with affinities across a 550-fold range, correlated well between FP and SPR assay formats for both human and mouse galectin-3. Increases in affinity for compounds binding to human galectin-3 were driven by changes in both kon and koff whilst for mouse galectin-3 this was primarily due to kon . The reduction in affinity observed between human to mouse galectin-3 was also comparable between assay formats. SPR has been shown to be a viable alternative to FP for early drug discovery screening and determining KD values. In addition, it can also provide early kinetic characterisation of small molecule galectin-3 glycomimetics with robust kon and koff values generated in a high throughput manner., Competing Interests: Declaration of Competing Interest The authors have been, or are currently, employees of Galecto Biotech AB or NovAliX., (Copyright © 2023 Galecto Biotech. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
22. Galectin-3: therapeutic targeting in liver disease.
- Author
-
Mackinnon AC, Tonev D, Jacoby B, Pinzani M, and Slack RJ
- Abstract
Introduction: The rising incidence of liver diseases is a worldwide healthcare concern. However, the therapeutic options to manage chronic inflammation and fibrosis, the processes at the basis of morbidity and mortality of liver diseases, are very limited. Galectin 3 (Gal-3) is a protein implicated in fibrosis in multiple organs. Several Gal-3 inhibitors are currently in clinical development., Areas Covered: This review describes our current understanding of the role of Gal-3 in chronic liver diseases, with special emphasis on fibrosis. Also, we review therapeutic advances based on Gal-3 inhibition, describing drug properties and their current status in clinical research., Expert Opinion: Currently, the known effects of Gal-3 point to a direct activation of the NLRP3 inflammasome leading to its activation in liver macrophages and activated macrophages play a key role in tissue fibrogenesis. However, more research is needed to elucidate the role of Gal-3 in the different activation pathways, dissecting the intracellular and extracellular mechanisms of Gal-3, and its role in pathogenesis. Gal-3 could be a target for early therapy of numerous hepatic diseases and, given the lack of therapeutic options for liver fibrosis, there is a strong pharmacologic potential for Gal-3-based therapies.
- Published
- 2023
- Full Text
- View/download PDF
23. Safety and pharmacokinetics of GB1211, an oral galectin-3 inhibitor: a single- and multiple-dose first-in-human study in healthy participants.
- Author
-
Aslanis V, Slack RJ, MacKinnon AC, McClinton C, Tantawi S, Gravelle L, Nilsson UJ, Leffler H, Brooks A, Khindri SK, Marshall RP, Pedersen A, Schambye H, and Zetterberg F
- Subjects
- Humans, Administration, Oral, Area Under Curve, Dose-Response Relationship, Drug, Double-Blind Method, Healthy Volunteers, Galectin 3 antagonists & inhibitors
- Abstract
Purpose: Galectin-3, a β-galactoside-binding lectin, plays a key role in several cellular pathways involved in chronic inflammation, heart disease and cancer. GB1211 is an orally bioavailable galectin-3 inhibitor, developed to be systemically active. We report safety and pharmacokinetics (PK) of GB1211 in healthy participants., Methods: This phase 1, double-blind, placebo-controlled, first-in-human study (NCT03809052) included a single ascending-dose phase (with a food-effect cohort) where participants across seven sequential cohorts were randomized 3:1 to receive oral GB1211 (5, 20, 50, 100, 200 or 400 mg) or placebo. In the multiple ascending-dose phase, participants received 50 or 100 mg GB1211 or placebo twice daily for 10 days. All doses were administered in the fasted state except in the food-effect cohort where doses were given 30 min after a high-fat meal., Results: All 78 participants received at least one GB1211 dose (n = 58) or placebo (n = 20) and completed the study. No safety concerns were identified. Following single and multiple oral doses under fasted conditions, maximum GB1211 plasma concentrations were reached at 1.75-4 h (median) post-dose; mean half-life was 11-16 h. There was a ~ twofold GB1211 accumulation in plasma with multiple dosing, with steady-state reached within 3 days; 30% of the administered dose was excreted in urine as unchanged drug. Absorption in the fed state was delayed by 2 h but systemic exposure was unaffected., Conclusion: GB1211 was well tolerated, rapidly absorbed, and displayed favorable PK, indicating a potential to treat multiple disease types. These findings support further clinical development of GB1211., Clinical Trial Registration: The study was registered with ClinicalTrials.gov (identifier: NCT03809052)., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
24. An Inhaled Galectin-3 Inhibitor in COVID-19 Pneumonitis: A Phase Ib/IIa Randomized Controlled Clinical Trial (DEFINE).
- Author
-
Gaughan EE, Quinn TM, Mills A, Bruce AM, Antonelli J, MacKinnon AC, Aslanis V, Li F, O'Connor R, Boz C, Mills R, Emanuel P, Burgess M, Rinaldi G, Valanciute A, Mills B, Scholefield E, Hardisty G, Findlay EG, Parker RA, Norrie J, Dear JW, Akram AR, Koch O, Templeton K, Dockrell DH, Walsh TS, Partridge S, Humphries D, Wang-Jairaj J, Slack RJ, Schambye H, Phung, Gravelle L, Lindmark B, Shankar-Hari M, Hirani N, Sethi T, and Dhaliwal K
- Subjects
- Humans, SARS-CoV-2, Galectin 3, Inflammation, Treatment Outcome, COVID-19
- Abstract
Rationale: High circulating galectin-3 is associated with poor outcomes in patients with coronavirus disease (COVID-19). We hypothesized that GB0139, a potent inhaled thiodigalactoside galectin-3 inhibitor with antiinflammatory and antifibrotic actions, would be safely and effectively delivered in COVID-19 pneumonitis. Objectives: Primary outcomes were safety and tolerability of inhaled GB0139 as an add-on therapy for patients hospitalized with COVID-19 pneumonitis. Methods: We present the findings of two arms of a phase Ib/IIa randomized controlled platform trial in hospitalized patients with confirmed COVID-19 pneumonitis. Patients received standard of care (SoC) or SoC plus 10 mg inhaled GB0139 twice daily for 48 hours, then once daily for up to 14 days or discharge. Measurements and Main Results: Data are reported from 41 patients, 20 of which were assigned randomly to receive GB0139. Primary outcomes: the GB0139 group experienced no treatment-related serious adverse events. Incidences of adverse events were similar between treatment arms (40 with GB0139 + SoC vs. 35 with SoC). Secondary outcomes: plasma GB0139 was measurable in all patients after inhaled exposure and demonstrated target engagement with decreased circulating galectin (overall treatment effect post-hoc analysis of covariance [ANCOVA] over days 2-7; P = 0.0099 vs. SoC). Plasma biomarkers associated with inflammation, fibrosis, coagulopathy, and major organ function were evaluated. Conclusions: In COVID-19 pneumonitis, inhaled GB0139 was well-tolerated and achieved clinically relevant plasma concentrations with target engagement. The data support larger clinical trials to determine clinical efficacy. Clinical trial registered with ClinicalTrials.gov (NCT04473053) and EudraCT (2020-002230-32).
- Published
- 2023
- Full Text
- View/download PDF
25. Discovery and Optimization of the First Highly Effective and Orally Available Galectin-3 Inhibitors for Treatment of Fibrotic Disease.
- Author
-
Zetterberg FR, MacKinnon A, Brimert T, Gravelle L, Johnsson RE, Kahl-Knutson B, Leffler H, Nilsson UJ, Pedersen A, Peterson K, Roper JA, Schambye H, Slack RJ, and Tantawi S
- Subjects
- Animals, Bleomycin pharmacology, Carbon Tetrachloride, Fibrosis, Liver Cirrhosis drug therapy, Liver Cirrhosis pathology, Lung, Mice, Thiogalactosides, Triazoles, Galectin 3 metabolism, Idiopathic Pulmonary Fibrosis chemically induced, Idiopathic Pulmonary Fibrosis drug therapy, Idiopathic Pulmonary Fibrosis pathology
- Abstract
Galectin-3 is a carbohydrate-binding protein central to regulating mechanisms of diseases such as fibrosis, cancer, metabolic, inflammatory, and heart disease. We recently found a high affinity (nM) thiodigalactoside GB0139 which currently is in clinical development (PhIIb) as an inhaled treatment of idiopathic pulmonary fibrosis. To enable treatment of systemically galectin-3 driven disease, we here present the first series of selective galectin-3 inhibitors combining high affinity (nM) with oral bioavailability. This was achieved by optimizing galectin-3 specificity and physical chemical parameters for a series of disubstituted monogalactosides. Further characterization showed that this class of compounds reduced profibrotic gene expression in liver myofibroblasts and displayed antifibrotic activity in CCl
4 -induced liver fibrosis and bleomycin-induced lung fibrosis mouse models. On the basis of the overall pharmacokinetic, pharmacodynamic, and safety profile, GB1211 was selected as the clinical candidate and is currently in phase IIa clinical trials as a potential therapy for liver cirrhosis and cancer.- Published
- 2022
- Full Text
- View/download PDF
26. Galectin-3 inhibitor GB0139 protects against acute lung injury by inhibiting neutrophil recruitment and activation.
- Author
-
Humphries DC, Mills R, Boz C, McHugh BJ, Hirani N, Rossi AG, Pedersen A, Schambye HT, Slack RJ, Leffler H, Nilsson UJ, Wang W, Sethi T, and Mackinnon AC
- Abstract
Rationale: Galectin-3 (Gal-3) drives fibrosis during chronic lung injury, however, its role in acute lung injury (ALI) remains unknown. Effective pharmacological therapies available for ALI are limited; identifying novel concepts in treatment is essential. GB0139 is a Gal-3 inhibitor currently under clinical investigation for the treatment of idiopathic pulmonary fibrosis. We investigate the role of Gal-3 in ALI and evaluate whether its inhibition with GB0139 offers a protective role. The effect of GB0139 on ALI was explored in vivo and in vitro. Methods: The pharmacokinetic profile of intra-tracheal ( i.t. ) GB0139 was investigated in C57BL/6 mice to support the daily dosing regimen. GB0139 (1-30 µg) was then assessed following acute i.t. lipopolysaccharide (LPS) and bleomycin administration. Histology, broncho-alveolar lavage fluid (BALf) analysis, and flow cytometric analysis of lung digests and BALf were performed. The impact of GB0139 on cell activation and apoptosis was determined in vitro using neutrophils and THP-1, A549 and Jurkat E6 cell lines . Results: GB0139 decreased inflammation severity via a reduction in neutrophil and macrophage recruitment and neutrophil activation. GB0139 reduced LPS-mediated increases in interleukin (IL)-6, tumor necrosis factor alpha (TNFα) and macrophage inflammatory protein-1-alpha. In vitro , GB0139 inhibited Gal-3-induced neutrophil activation, monocyte IL-8 secretion, T cell apoptosis and the upregulation of pro-inflammatory genes encoding for IL-8, TNFα, IL-6 in alveolar epithelial cells in response to mechanical stretch. Conclusion: These data indicate that Gal-3 adopts a pro-inflammatory role following the early stages of lung injury and supports the development of GB0139, as a potential treatment approach in ALI., Competing Interests: AM and TS report personal fees from Galecto Biotech, outside the submitted work and AP, RS and HS report personal fees from Galecto Biotech, outside the submitted work. UN and HL are consultants of and shareholders in Galecto Biotech. NH received grants from Galecto Biotech. AR, BM, CB, DH, RM, WW declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. Galecto Biotech AB is fully the owner of granted patents US9243021, US9580456, US9688713, US7700763, US86977862, US10369136, US10307403, US10799482, EP2914269, EP2297174, EP2679595, CA2794066, CA2724064, IN279934, CN102066393, CN103497228, JP6863984 and pending patent applications WO2020260351, EP16809870, CA3004632, CN2016800710578, JP2021062805, IN201817023621, IN2573/DELNP/2015, US17/221201., (Copyright © 2022 Humphries, Mills, Boz, McHugh, Hirani, Rossi, Pedersen, Schambye, Slack, Leffler, Nilsson, Wang, Sethi and Mackinnon.)
- Published
- 2022
- Full Text
- View/download PDF
27. Emerging therapeutic opportunities for integrin inhibitors.
- Author
-
Slack RJ, Macdonald SJF, Roper JA, Jenkins RG, and Hatley RJD
- Subjects
- Animals, Drug Discovery methods, Humans, Protein Binding drug effects, Integrins antagonists & inhibitors, Integrins metabolism, Small Molecule Libraries pharmacology, Small Molecule Libraries therapeutic use
- Abstract
Integrins are cell adhesion and signalling proteins crucial to a wide range of biological functions. Effective marketed treatments have successfully targeted integrins αIIbβ3, α4β7/α4β1 and αLβ2 for cardiovascular diseases, inflammatory bowel disease/multiple sclerosis and dry eye disease, respectively. Yet, clinical development of others, notably within the RGD-binding subfamily of αv integrins, including αvβ3, have faced significant challenges in the fields of cancer, ophthalmology and osteoporosis. New inhibitors of the related integrins αvβ6 and αvβ1 have recently come to the fore and are being investigated clinically for the treatment of fibrotic diseases, including idiopathic pulmonary fibrosis and nonalcoholic steatohepatitis. The design of integrin drugs may now be at a turning point, with opportunities to learn from previous clinical trials, to explore new modalities and to incorporate new findings in pharmacological and structural biology. This Review intertwines research from biological, clinical and medicinal chemistry disciplines to discuss historical and current RGD-binding integrin drug discovery, with an emphasis on small-molecule inhibitors of the αv integrins., (© 2021. Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
28. Pharmacological characterisation of GSK3335103, an oral αvβ6 integrin small molecule RGD-mimetic inhibitor for the treatment of fibrotic disease.
- Author
-
Wilkinson AL, John AE, Barrett JW, Gower E, Morrison VS, Man Y, Pun KT, Roper JA, Luckett JC, Borthwick LA, Barksby BS, Burgoyne RA, Barnes R, Fisher AJ, Procopiou PA, Hatley RJD, Barrett TN, Marshall RP, Macdonald SJF, Jenkins RG, and Slack RJ
- Subjects
- Administration, Oral, Animals, Antifibrotic Agents chemistry, Antifibrotic Agents therapeutic use, Antigens, Neoplasm chemistry, Antigens, Neoplasm metabolism, Biological Availability, Bleomycin administration & dosage, Bleomycin toxicity, Cells, Cultured, Disease Models, Animal, Epithelial Cells drug effects, Epithelial Cells pathology, Humans, Integrins chemistry, Integrins metabolism, Lung drug effects, Lung pathology, Lysosomes metabolism, Male, Mice, Oligopeptides chemistry, Primary Cell Culture, Proteolysis drug effects, Pulmonary Fibrosis chemically induced, Pulmonary Fibrosis pathology, Transforming Growth Factor beta metabolism, Antifibrotic Agents pharmacology, Integrins antagonists & inhibitors, Pulmonary Fibrosis drug therapy
- Abstract
Fibrosis is the formation of scar tissue due to injury or long-term inflammation and is a leading cause of morbidity and mortality. Activation of the pro-fibrotic cytokine transforming growth factor-β (TGFβ) via the alpha-V beta-6 (αvβ6) integrin has been identified as playing a key role in the development of fibrosis. Therefore, a drug discovery programme to identify an orally bioavailable small molecule αvβ6 arginyl-glycinyl-aspartic acid (RGD)-mimetic was initiated. As part of a medicinal chemistry programme GSK3335103 was identified and profiled in a range of pre-clinical in vitro and in vivo systems. GSK3335103 was shown to bind to the αvβ6 with high affinity and demonstrated fast binding kinetics. In primary human lung epithelial cells, GSK3335103-induced concentration- and time-dependent internalisation of αvβ6 with a rapid return of integrin to the cell surface observed after washout. Following sustained engagement of the αvβ6 integrin in vitro, lysosomal degradation was induced by GSK3335103. GSK3335103 was shown to engage with the αvβ6 integrin and inhibit the activation of TGFβ in both ex vivo IPF tissue and in a murine model of bleomycin-induced lung fibrosis, as measured by αvβ6 engagement, TGFβ signalling and collagen deposition, with a prolonged duration of action observed in vivo. In summary, GSK3335103 is a potent αvβ6 inhibitor that attenuates TGFβ signalling in vitro and in vivo with a well-defined pharmacokinetic/pharmacodynamic relationship. This translates to a significant reduction of collagen deposition in vivo and therefore GSK3335103 represents a potential novel oral therapy for fibrotic disorders., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
29. Target inhibition of galectin-3 by inhaled TD139 in patients with idiopathic pulmonary fibrosis.
- Author
-
Hirani N, MacKinnon AC, Nicol L, Ford P, Schambye H, Pedersen A, Nilsson UJ, Leffler H, Sethi T, Tantawi S, Gravelle L, Slack RJ, Mills R, Karmakar U, Humphries D, Zetterberg F, Keeling L, Paul L, Molyneaux PL, Li F, Funston W, Forrest IA, Simpson AJ, Gibbons MA, and Maher TM
- Subjects
- Double-Blind Method, Humans, Lung, Galectin 3, Idiopathic Pulmonary Fibrosis
- Abstract
Galectin (Gal)-3 is a profibrotic β-galactoside-binding lectin that plays a key role in the pathogenesis of idiopathic pulmonary fibrosis (IPF) and IPF exacerbations. TD139 is a novel and potent small-molecule inhibitor of Gal-3.A randomised, double-blind, multicentre, placebo-controlled, phase 1/2a study was conducted to assess the safety, tolerability, pharmacokinetics and pharmacodynamics of inhaled TD139 in 36 healthy subjects and 24 patients with IPF. Six dose cohorts of six healthy subjects were evaluated (4:2 TD139:placebo ratio) with single doses of TD139 (0.15-50 mg) and three dose cohorts of eight patients with IPF (5:3 TD139:placebo ratio) with once-daily doses of TD139 (0.3-10 mg) for 14 days.Inhaled TD139 was well tolerated with no significant treatment-related side-effects. TD139 was rapidly absorbed, with mean time taken to reach maximum plasma concentration ( C
max ) values ranging from 0.6 to 3 h and a plasma half-life ( T1/2 ) of 8 h. The concentration of TD139 in the lung was >567-fold higher than in the blood, with systemic exposure predicting exposure in the target compartment. Gal-3 expression on alveolar macrophages was reduced in the 3 and 10 mg dose groups compared with placebo, with a concentration-dependent inhibition demonstrated. Inhibition of Gal-3 expression in the lung was associated with reductions in plasma biomarkers centrally relevant to IPF pathobiology (platelet-derived growth factor-BB, plasminogen activator inhibitor-1, Gal-3, CCL18 and YKL-40).TD139 is safe and well tolerated in healthy subjects and IPF patients. It was shown to suppress Gal-3 expression on bronchoalveolar lavage macrophages and, in a concerted fashion, decrease plasma biomarkers associated with IPF progression., Competing Interests: Conflict of interest: N. Hirani reports grants from Galecto Biotech, during the conduct of the study. Conflict of interest: A.C. MacKinnon reports personal fees from Galecto Biotech, outside the submitted work; and has a patent CA2,794,066 issued, a patent US13/832,672 issued and a patent WO/2014/067986 pending (all patents are fully owned by Galecto Biotech). Conflict of interest: L. Nicol reports grants from Galecto Biotech, during the conduct of the study; personal fees for lectures from Boehringer Ingelheim, outside the submitted work. Conflict of interest: P. Ford reports personal fees and nonfinancial support from Galecto, during the conduct of the study; and has a patent TD139 issued. Conflict of interest: H. Schambye reports personal fees from Galecto Inc, outside the submitted work; and has a patent WO/2016/180483 pending (fully owned by Galecto Biotech). Conflict of interest: A. Pedersen reports personal fees from Galecto Biotech, outside the submitted work. Conflict of interest: U.J. Nilsson has a patent CA2,794,066 issued, a patent US13/832,672 issued, a patent WO/2014/067986 pending, a patent WO/2005/113569 pending and a patent WO/2009/139719 pending (all patents are fully owned by Galecto Biotech). Conflict of interest: H. Leffler has a patent CA2,794,066 issued, a patent US13/832,672 issued, a patent WO/2014/067986 pending and a patent WO/2005/113569 pending (all patents are fully owned by Galecto Biotech). Conflict of interest: T. Sethi reports personal fees from Galecto Biotech, outside the submitted work; and has a patent CA2,794,066 issued, a patent US13/832,672 issued and a patent WO/2014/067986 pending (patents are fully owned by Galecto Biotech). Conflict of interest: S. Tantawi reports personal fees from Galecto Biotech, outside the submitted work. Conflict of interest: L. Gravelle reports personal fees from Galecto Biotech, outside the submitted work; and has a patent WO/2017/103109 pending (fully owned by Galecto Biotech). Conflict of interest: R.J. Slack reports personal fees from Galecto Biotech, outside the submitted work. Conflict of interest: R. Mills has nothing to disclose. Conflict of interest: U. Karmakar has nothing to disclose. Conflict of interest: D. Humphries has nothing to disclose. Conflict of interest: F. Zetterberg reports personal fees from Galecto Biotech, outside the submitted work. Conflict of interest: L. Keeling has nothing to disclose. Conflict of interest: L. Paul has nothing to disclose. Conflict of interest: P.L. Molyneaux has, via his institution, received industry-academic funding from AstraZeneca and has received speaker and consultancy fees from Boehringer Ingelheim and Hoffman-La Roche, outside the submitted work. Conflict of interest: F. Li has nothing to disclose. Conflict of interest: W. Funston has nothing to disclose. Conflict of interest: I.A. Forrest reports personal fees for consultancy and meeting attendance from Boehringer Ingelheim, personal fees for lectures and meeting attendance from Roche Ltd, outside the submitted work. Conflict of interest: A.J. Simpson has nothing to disclose. Conflict of interest: M.A. Gibbons has nothing to disclose. Conflict of interest: T.M. Maher has, via his institution, received industry-academic funding from AstraZeneca and GlaxoSmithKline R&D, and has received consultancy or speaker fees from AstraZeneca, Bayer, Blade Therapeutics, Boehringer Ingelheim, Bristol-Myers Squibb, Galapagos, GlaxoSmithKline R&D, Indalo, Novartis, Pliant, Respivant, Roche and Samumed., (Copyright ©ERS 2021.)- Published
- 2021
- Full Text
- View/download PDF
30. Downregulation of the α v β 6 Integrin via RGD Engagement Is Affinity and Time Dependent.
- Author
-
Roper JA, Wilkinson AL, Gower E, and Slack RJ
- Subjects
- Antigens, Neoplasm chemistry, Binding Sites, Cells, Cultured, Epithelial Cells drug effects, Epithelial Cells metabolism, Humans, Integrins chemistry, Kinetics, Lysosomes metabolism, Oligopeptides metabolism, Phenylpropionates pharmacology, Protein Binding, Proteolysis, Respiratory Mucosa cytology, Transforming Growth Factor beta metabolism, Antigens, Neoplasm metabolism, Down-Regulation, Integrins metabolism, Transforming Growth Factor beta antagonists & inhibitors
- Abstract
The arginyl-glycinyl-aspartic acid (RGD) integrin alpha-v beta-6 ( α v β 6) has been identified as playing a key role in the activation of transforming growth factor- β (TGF β ) that is hypothesized to be pivotal in the development of fibrosis and other diseases. In this study, α v β 6 small molecule inhibitors were characterized in a range of in vitro systems to determine affinity, kinetics, and duration of TGF β inhibition. High α v β 6 binding affinity was shown to be correlated with slow dissociation kinetics. Compound 1 (high α v β 6 affinity, slow dissociation) and SC-68448 (low α v β 6 affinity, fast dissociation) induced concentration- and time-dependent internalization of α v β 6 in normal human bronchial epithelial (NHBE) cells. After washout, the α v β 6 cell surface repopulation was faster for SC-68448 compared with compound 1 In addition, α v β 6-dependent release of active TGF β from NHBE cells was inhibited by compound 1 and SC-68448. After washout of SC-68448, release of active TGF β was restored, whereas after washout of compound 1 the inhibition of TGF β activation was maintained and only reversible in the presence of a lysosomal inhibitor (chloroquine). However, SC-68448 was able to reduce total levels of α v β 6 in NHBE cells if present continuously. These observations suggest α v β 6 can be degraded after high affinity RGD binding that sorts the integrin for lysosomal degradation after internalization, likely due to sustained engagement as a result of slow dissociation kinetics. In addition, the α v β 6 integrin can also be downregulated after sustained engagement of the RGD binding site with low affinity ligands that do not sort the integrin for immediate lysosomal degradation. SIGNIFICANCE STATEMENT: The fate of RGD integrin after ligand binding has not been widely investigated. Using the αvβ6 integrin as a case study, we have demonstrated that RGD-induced downregulation of αvβ6 is both affinity and time dependent. High affinity ligands induced downregulation via lysosomal degradation, likely due to slow dissociation, whereas sustained low affinity ligand engagement was only able to decrease αvβ6 expression over longer periods of time. Our study provides a potential unique mechanism for obtaining duration of action for drugs targeting integrins., (Copyright © 2021 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2021
- Full Text
- View/download PDF
31. The therapeutic potential of galectin-3 inhibition in fibrotic disease.
- Author
-
Slack RJ, Mills R, and Mackinnon AC
- Subjects
- Animals, Blood Proteins antagonists & inhibitors, Fibrosis metabolism, Fibrosis pathology, Galectins metabolism, Humans, Inflammation metabolism, Inflammation pathology, Endocytosis, Fibrosis drug therapy, Galectins antagonists & inhibitors, Inflammation prevention & control
- Abstract
Galectin-3 is a beta-galactoside-binding mammalian lectin and part of the 15 member galectin family that are evolutionarily highly conserved. It is the only chimeric protein with a C-terminal carbohydrate recognition domain (CRD) linked to a proline, glycine, and tyrosine rich additional N-terminal domain. Galectin-3 binds several cell surface glycoproteins via its CRD domain as well as undergoing oligomerization, via binding at the N-terminal or the CRD, resulting in the formation of a galectin-3 lattice on the cell surface. The galectin-3 lattice has been regarded as being a crucial mechanism whereby extracellular galectin-3 modulates cellular signalling by prolonging retention time or retarding lateral movement of cell surface receptors in the plasma membrane. As such galectin-3 can regulate various cellular functions such as diffusion, compartmentalization and endocytosis of plasma membrane glycoproteins and glycolipids and the functionality of membrane receptors. In multiple models of organ fibrosis, it has been demonstrated that galectin-3 is potently pro-fibrotic and modulates the activity of fibroblasts and macrophages in chronically inflamed organs. Increased galectin-3 expression also activates myofibroblasts resulting in scar formation and may therefore impact common fibrotic pathways leading to fibrosis in multiple organs. Over the last decade there has been a marked increase in the scientific literature investigating galectin-3 in a range of fibrotic diseases as well as the clinical development of new galectin-3 inhibitors. In this review we will examine the role of galectin-3 in fibrosis, the therapeutic strategies for inhibiting galectin-3 in fibrotic disease and the clinical landscape to date., (Copyright © 2020 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
32. Translational pharmacology of an inhaled small molecule αvβ6 integrin inhibitor for idiopathic pulmonary fibrosis.
- Author
-
John AE, Graves RH, Pun KT, Vitulli G, Forty EJ, Mercer PF, Morrell JL, Barrett JW, Rogers RF, Hafeji M, Bibby LI, Gower E, Morrison VS, Man Y, Roper JA, Luckett JC, Borthwick LA, Barksby BS, Burgoyne RA, Barnes R, Le J, Flint DJ, Pyne S, Habgood A, Organ LA, Joseph C, Edwards-Pritchard RC, Maher TM, Fisher AJ, Gudmann NS, Leeming DJ, Chambers RC, Lukey PT, Marshall RP, Macdonald SJF, Jenkins RG, and Slack RJ
- Subjects
- Administration, Inhalation, Animals, Antigens, Neoplasm metabolism, Bleomycin toxicity, Butyrates administration & dosage, Butyrates metabolism, Butyrates pharmacokinetics, Collagen metabolism, Disease Models, Animal, Epithelial Cells drug effects, Humans, Idiopathic Pulmonary Fibrosis chemically induced, Idiopathic Pulmonary Fibrosis pathology, Integrins metabolism, Male, Mice, Inbred C57BL, Molecular Docking Simulation, Naphthyridines administration & dosage, Naphthyridines metabolism, Naphthyridines pharmacokinetics, Pyrazoles administration & dosage, Pyrazoles metabolism, Pyrazoles pharmacokinetics, Pyrrolidines administration & dosage, Pyrrolidines metabolism, Pyrrolidines pharmacokinetics, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology, Tomography, Emission-Computed, Single-Photon, Transforming Growth Factor beta metabolism, Translational Research, Biomedical, Butyrates pharmacology, Idiopathic Pulmonary Fibrosis drug therapy, Integrins antagonists & inhibitors, Naphthyridines pharmacology, Pyrazoles pharmacology, Pyrrolidines pharmacology
- Abstract
The αvβ6 integrin plays a key role in the activation of transforming growth factor-β (TGFβ), a pro-fibrotic mediator that is pivotal to the development of idiopathic pulmonary fibrosis (IPF). We identified a selective small molecule αvβ6 RGD-mimetic, GSK3008348, and profiled it in a range of disease relevant pre-clinical systems. To understand the relationship between target engagement and inhibition of fibrosis, we measured pharmacodynamic and disease-related end points. Here, we report, GSK3008348 binds to αvβ6 with high affinity in human IPF lung and reduces downstream pro-fibrotic TGFβ signaling to normal levels. In human lung epithelial cells, GSK3008348 induces rapid internalization and lysosomal degradation of the αvβ6 integrin. In the murine bleomycin-induced lung fibrosis model, GSK3008348 engages αvβ6, induces prolonged inhibition of TGFβ signaling and reduces lung collagen deposition and serum C3M, a marker of IPF disease progression. These studies highlight the potential of inhaled GSK3008348 as an anti-fibrotic therapy.
- Published
- 2020
- Full Text
- View/download PDF
33. Preclinical evaluation of [ 18 F]FB-A20FMDV2 as a selective marker for measuring α V β 6 integrin occupancy using positron emission tomography in rodent lung.
- Author
-
Onega M, Parker CA, Coello C, Rizzo G, Keat N, Ramada-Magalhaes J, Moz S, Tang SP, Plisson C, Wells L, Ashworth S, Slack RJ, Vitulli G, Wilson FJ, Gunn R, Lukey PT, and Passchier J
- Subjects
- Animals, Antigens, Neoplasm, Integrin beta Chains, Lung diagnostic imaging, Positron-Emission Tomography, Rats, Rats, Sprague-Dawley, Tissue Distribution, Integrins metabolism, Rodentia metabolism
- Abstract
Purpose: Integrin α
v β6 belongs to the RGD subset of the integrin family, and its expression levels are a prognostic and theranostic factor in some types of cancer and pulmonary fibrosis. This paper describes the GMP radiolabelling of the synthetic 20 amino acid peptide A20FMDV2 (NAVPNLRGDLQVLAQKVART), derived from the foot-and-mouth disease virus, and characterises the use of [18 F]FB-A20FMDV2 as a high affinity, specific and selective PET radioligand for the quantitation and visualisation of αv β6 in rodent lung to support human translational studies., Methods: The synthesis of [18 F]FB-A20FMDV2 was performed using a fully automated and GMP-compliant process. Sprague-Dawley rats were used to perform homologous (unlabelled FB-A20FMDV2) and heterologous (anti-αv β6 antibody 8G6) blocking studies. In order to generate a dosimetry estimate, tissue residence times were generated, and associated tissue exposure and effective dose were calculated using the Organ Level Internal Dose Assessment/Exponential Modelling (OLINDA/EXM) software., Results: [18 F]FB-A20FMDV2 synthesis was accomplished in 180 min providing ~800 MBq of [18 F]FB-A20FMDV2 with a molar activity of up to 150 GBq/μmol and high radiochemical purity (> 97%). Following i.v. administration to rats, [18 F]FB-A20FMDV2 was rapidly metabolised with intact radiotracer representing 5% of the total radioactivity present in rat plasma at 30 min. For the homologous and heterologous block in rats, lung-to-heart SUV ratios at 30-60 min post-administration of [18 F]FB-A20FMDV2 were reduced by 38.9 ± 6.9% and 56 ± 19.2% for homologous and heterologous block, respectively. Rodent biodistribution and dosimetry calculations using OLINDA/EXM provided a whole body effective dose in humans 33.5 μSv/MBq., Conclusion: [18 F]FB-A20FMDV2 represents a specific and selective PET ligand to measure drug-associated αvβ6 integrin occupancy in lung. The effective dose, extrapolated from rodent data, is in line with typical values for compounds labelled with fluorine-18 and combined with the novel fully automated and GMP-compliant synthesis and allows for clinical use in translational studies.- Published
- 2020
- Full Text
- View/download PDF
34. A positron emission tomography imaging study to confirm target engagement in the lungs of patients with idiopathic pulmonary fibrosis following a single dose of a novel inhaled αvβ6 integrin inhibitor.
- Author
-
Maher TM, Simpson JK, Porter JC, Wilson FJ, Chan R, Eames R, Cui Y, Siederer S, Parry S, Kenny J, Slack RJ, Sahota J, Paul L, Saunders P, Molyneaux PL, Lukey PT, Rizzo G, Searle GE, Marshall RP, Saleem A, Kang'ombe AR, Fairman D, Fahy WA, and Vahdati-Bolouri M
- Subjects
- Administration, Inhalation, Aged, Antigens, Neoplasm, Bayes Theorem, Butyrates administration & dosage, Butyrates pharmacokinetics, Double-Blind Method, Endpoint Determination, Female, Humans, Idiopathic Pulmonary Fibrosis diagnostic imaging, Male, Naphthyridines administration & dosage, Naphthyridines pharmacokinetics, Nebulizers and Vaporizers, Positron-Emission Tomography, Pyrazoles administration & dosage, Pyrazoles pharmacokinetics, Pyrrolidines administration & dosage, Pyrrolidines pharmacokinetics, Treatment Outcome, Butyrates therapeutic use, Idiopathic Pulmonary Fibrosis drug therapy, Integrins antagonists & inhibitors, Naphthyridines therapeutic use, Pyrazoles therapeutic use, Pyrrolidines therapeutic use, Tidal Volume drug effects
- Abstract
Background: Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive lung disease with poor prognosis and a significant unmet medical need. This study evaluated the safety, pharmacokinetics (PK) and target engagement in the lungs, of GSK3008348, a novel inhaled alpha-v beta-6 (αvβ6) integrin inhibitor, in participants with IPF., Methods: This was a phase 1b, randomised, double-blind (sponsor unblind) study, conducted in the UK (two clinical sites, one imaging unit) between June 2017 and July 2018 (NCT03069989). Participants with a definite or probable diagnosis of IPF received a single nebulised dose of 1000 mcg GSK3008348 or placebo (ratio 5:2) in two dosing periods. In period 1, safety and PK assessments were performed up to 24 h post-dose; in period 2, after a 7-day to 28-day washout, participants underwent a total of three positron emission tomography (PET) scans: baseline, Day 1 (~ 30 min post-dosing) and Day 2 (~ 24 h post-dosing), using a radiolabelled αvβ6-specific ligand, [
18 F]FB-A20FMDV2. The primary endpoint was whole lung volume of distribution (VT ), not corrected for air volume, at ~ 30 min post-dose compared with pre-dose. The study success criterion, determined using Bayesian analysis, was a posterior probability (true % reduction in VT > 0%) of ≥80%., Results: Eight participants with IPF were enrolled and seven completed the study. Adjusted posterior median reduction in uncorrected VT at ~ 30 min after GSK3008348 inhalation was 20% (95% CrI: - 9 to 42%). The posterior probability that the true % reduction in VT > 0% was 93%. GSK3008348 was well tolerated with no reports of serious adverse events or clinically significant abnormalities that were attributable to study treatment. PK was successfully characterised showing rapid absorption followed by a multiphasic elimination., Conclusions: This study demonstrated engagement of the αvβ6 integrin target in the lung following nebulised dosing with GSK3008348 to participants with IPF. To the best of our knowledge this is the first time a target-specific PET radioligand has been used to assess target engagement in the lung, not least for an inhaled drug., Trial Registration: clinicaltrials.gov: NCT03069989; date of registration: 3 March 2017.- Published
- 2020
- Full Text
- View/download PDF
35. Profile of a Highly Selective Quaternized Pyrrolidine Betaine α v β 6 Integrin Inhibitor-(3 S )-3-(3-(3,5-Dimethyl-1 H -pyrazol-1-yl)phenyl)-4-((1 S and 1 R ,3 R )-1-methyl-3-(2-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)ethyl)pyrrolidin-1-ium-1-yl)butanoate Synthesized by Stereoselective Methylation.
- Author
-
Barrett TN, Taylor JA, Barker D, Procopiou PA, Thompson JDF, Barrett J, Le J, Lynn SM, Pogany P, Pratley C, Pritchard JM, Roper JA, Rowedder JE, Slack RJ, Vitulli G, Macdonald SJF, and Kerr WJ
- Subjects
- Animals, Antigens, Neoplasm chemistry, Antigens, Neoplasm metabolism, Betaine chemistry, Betaine pharmacokinetics, Cells, Cultured, Crystallography, X-Ray, Hepatocytes cytology, Hepatocytes drug effects, Hepatocytes metabolism, Humans, Integrins chemistry, Integrins metabolism, Methylation, Models, Chemical, Molecular Docking Simulation, Molecular Structure, Protein Binding, Protein Conformation, Quaternary Ammonium Compounds chemistry, Quaternary Ammonium Compounds pharmacokinetics, Rats, Stereoisomerism, Betaine pharmacology, Integrins antagonists & inhibitors, Pyrrolidines chemistry, Quaternary Ammonium Compounds pharmacology
- Abstract
A quaternary ammonium betaine 7 is described which shows exceptional potency and selectivity (1.4 to >3 logs) for the α
v β6 integrin receptor over the other αv integrins as determined in cell adhesion assays. 7 is prepared by remarkably stereoselective methylation, the origins of which are discussed. The chemical, biological, physicochemical, and pharmacokinetic properties of 7 and its docking into αv β6 are described along with related analogues.- Published
- 2019
- Full Text
- View/download PDF
36. The Design of Potent, Selective and Drug-Like RGD αvβ1 Small-Molecule Inhibitors Derived from non-RGD α4β1 Antagonists.
- Author
-
Hatley RJD, Barrett TN, Slack RJ, Watson ME, Baillache DJ, Gruszka A, Washio Y, Rowedder JE, Pogány P, Pal S, and Macdonald SJF
- Subjects
- Animals, Binding Sites, Drug Design, Drug Stability, Humans, Ligands, Liver metabolism, Male, Phenylalanine chemical synthesis, Phenylalanine metabolism, Rats, Sprague-Dawley, Receptors, Vitronectin chemistry, Receptors, Vitronectin metabolism, Urea chemical synthesis, Urea metabolism, Phenylalanine analogs & derivatives, Receptors, Vitronectin antagonists & inhibitors, Urea analogs & derivatives
- Abstract
Up to 45 % of deaths in developed nations can be attributed to chronic fibroproliferative diseases, highlighting the need for effective therapies. The RGD (Arg-Gly-Asp) integrin αvβ1 was recently investigated for its role in fibrotic disease, and thus warrants therapeutic targeting. Herein we describe the identification of non-RGD hit small-molecule αvβ1 inhibitors. We show that αvβ1 activity is embedded in a range of published α4β1 (VLA-4) ligands; we also demonstrate how a non-RGD integrin inhibitor (of α4β1 in this case) was converted into a potent non-zwitterionic RGD integrin inhibitor (of αvβ1 in this case). We designed urea ligands with excellent selectivity over α4β1 and the other αv integrins (αvβ3, αvβ5, αvβ6, αvβ8). In silico docking models and density functional theory (DFT) calculations aided the discovery of the lead urea series., (© 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2019
- Full Text
- View/download PDF
37. The effect of divalent metal cations on the αv integrin binding site is ligand and integrin specific.
- Author
-
Hall ER and Slack RJ
- Subjects
- Animals, Binding Sites drug effects, Binding Sites physiology, CHO Cells, Cations, Divalent analysis, Cations, Divalent pharmacology, Cricetinae, Cricetulus, Dose-Response Relationship, Drug, Humans, Integrin alphaV analysis, Integrins analysis, Integrins metabolism, Ligands, Cations, Divalent metabolism, Integrin alphaV metabolism, Radioligand Assay methods
- Abstract
The binding of orthosteric ligands to integrins requires the presence of divalent metal cations bound to metal ion-binding sites located in the I domains of the integrin α and β subunits. In this study the influence of the type and concentration of divalent metal cation present was investigated on a single arginyl-glycinyl-aspartic acid (RGD) ligand across the αv integrin sub-family and single αv integrin (αvβ6) with different ligands. These relationships were determined using radioligand binding studies completed with [
3 H] ligands and purified αv integrin protein preparations. The binding of [3 H]compound 1 to the RGD site on individual αv integrins demonstrated a unique profile in relation to the type and concentration of divalent metal cation present. The use of physiological concentrations of Mg2+ and Ca2+ in simulated lung fluid altered the αv integrin selectivity profile of [3 H]compound 1 in terms of affinity and the level of receptor occupancy. In addition, different RGD ligands for the αvβ6 integrin behaved differently under the same divalent metal cation conditions. In conclusion, this study demonstrates the need to determine the individual relationship between RGD ligands and the integrins they may engage in vivo, especially when determining selectivity profiles for potential RGD-mimetic small molecule therapeutics, with organ and disease state also considered., (Copyright © 2018 GlaxoSmithKline Plc. Published by Elsevier Masson SAS.. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
38. Pharmacological characterisation of a tool αvβ1 integrin small molecule RGD-mimetic inhibitor.
- Author
-
Wilkinson AL, Barrett JW, and Slack RJ
- Subjects
- Animals, Humans, Male, Mice, Mice, Inbred C57BL, Peptidomimetics chemistry, Peptidomimetics metabolism, Peptidomimetics pharmacokinetics, Receptors, Vitronectin chemistry, Oligopeptides chemistry, Peptidomimetics pharmacology, Receptors, Vitronectin metabolism
- Abstract
Compound 8 is a selective αvβ1 small molecule inhibitor that has been used in pre-clinical studies to identify and characterise the αvβ1 integrin as a potential target in fibrotic disease. In this study we further investigated the selectivity and pharmacokinetics of compound 8 to determine a link between the levels of αvβ1 engagement required to achieve in vivo pharmacodynamic efficacy. The selectivity of compound 8 for the arginyl-glycinyl-aspartic acid and β1 integrins was measured using purified integrin protein preparations in radioligand binding studies with both labelled ([
3 H]compound 8) and unlabelled versions. The pharmacokinetic profile of compound 8 was completed in in vitro blood protein binding assays and in in vivo studies using male C57BL/6 mouse following i.v. dosing. The high selectivity of compound 8 for αvβ1 over the other αv integrins was confirmed, however a reduced selectivity was demonstrated for the β1 integrin family, with high affinity observed for α4β1 (comparable to αvβ1), moderate affinity for α2β1, α3β1 and α8β1, and low affinity for α5β1 and α9β1. Compound 8 was shown to be cleared quickly from the blood with a short half-life of 0.5 h. In conclusion, the data in this study suggest that compound 8 has the potential to engage a number of integrins in vivo beyond αvβ1, that raises a degree of uncertainty regarding its mechanism of action in models of fibrotic disease., (Copyright © 2018 Elsevier B.V. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
39. Discovery of ( S)-3-(3-(3,5-Dimethyl-1 H-pyrazol-1-yl)phenyl)-4-(( R)-3-(2-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)ethyl)pyrrolidin-1-yl)butanoic Acid, a Nonpeptidic α v β 6 Integrin Inhibitor for the Inhaled Treatment of Idiopathic Pulmonary Fibrosis.
- Author
-
Procopiou PA, Anderson NA, Barrett J, Barrett TN, Crawford MHJ, Fallon BJ, Hancock AP, Le J, Lemma S, Marshall RP, Morrell J, Pritchard JM, Rowedder JE, Saklatvala P, Slack RJ, Sollis SL, Suckling CJ, Thorp LR, Vitulli G, and Macdonald SJF
- Subjects
- Animals, Antigens, Neoplasm, Cell Adhesion, Dogs, Humans, Lung metabolism, Male, Mice, Models, Molecular, Molecular Structure, Protein Conformation, Rats, Rats, Wistar, Structure-Activity Relationship, Tissue Distribution, Drug Discovery, Idiopathic Pulmonary Fibrosis drug therapy, Integrins antagonists & inhibitors, Lung drug effects, Pyrazoles chemistry
- Abstract
A series of 3-aryl(pyrrolidin-1-yl)butanoic acids were synthesized using a diastereoselective route, via a rhodium catalyzed asymmetric 1,4-addition of arylboronic acids in the presence of ( R)-BINAP to a crotonate ester to provide the ( S) absolute configuration for the major product. A variety of aryl substituents including morpholine, pyrazole, triazole, imidazole, and cyclic ether were screened in cell adhesion assays for affinity against α
v β1 , αv β3 , αv β5 , αv β6 , and αv β8 integrins. Numerous analogs with high affinity and selectivity for the αv β6 integrin were identified. The analog ( S)-3-(3-(3,5-dimethyl-1 H-pyrazol-1-yl)phenyl)-4-(( R)-3-(2-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)ethyl)pyrrolidin-1-yl)butanoic acid hydrochloride salt was found to have very high affinity for αv β6 integrin in a radioligand binding assay (p Ki = 11), a long dissociation half-life (7 h), very high solubility in saline at pH 7 (>71 mg/mL), and pharmacokinetic properties commensurate with inhaled dosing by nebulization. It was selected for further clinical investigation as a potential therapeutic agent for the treatment of idiopathic pulmonary fibrosis.- Published
- 2018
- Full Text
- View/download PDF
40. A Microdose PET Study of the Safety, Immunogenicity, Biodistribution, and Radiation Dosimetry of 18 F-FB-A20FMDV2 for Imaging the Integrin α v β 6 .
- Author
-
Keat N, Kenny J, Chen K, Onega M, Garman N, Slack RJ, Parker CA, Lumbers RT, Hallett W, Saleem A, Passchier J, and Lukey PT
- Subjects
- Aged, Amino Acid Sequence, Female, Foot-and-Mouth Disease Virus, Humans, Male, Middle Aged, Peptide Fragments adverse effects, Peptide Fragments chemistry, Radiometry, Tissue Distribution, Viral Proteins chemistry, Antigens, Neoplasm metabolism, Fluorine Radioisotopes, Integrins metabolism, Peptide Fragments immunology, Peptide Fragments pharmacokinetics, Positron-Emission Tomography methods, Radiation Dosage, Safety
- Abstract
The α
v β6 integrin is involved in the pathogenesis of cancer and fibrosis. A radiolabeled 20-amino-acid αv β6 -binding peptide, derived from the foot and mouth virus (NAVPNLRGDLQVLAQKVART [A20FMDV2]), has been developed to image αv β6 levels preclinically. This study was designed to translate these findings into a clinical PET imaging protocol to measure the expression of αv β6 in humans. Methods: Preclinical toxicology was undertaken, and a direct immunoassay was developed for 4-fluorobenzamide (FB)-A20FMDV2. Four healthy human subjects (2 male and 2 female) received a single microdose of18 F-FB-A20FMDV2 followed by a multibed PET scan of the whole body over more than 3 h. Results: There were no findings in the preclinical toxicology assessments, and no anti-A20FMDV2 antibodies were detected before or after dosing with the PET ligand. The mean and SD of the administered mass of18 F-FB-A20FMDV2 was 8.7 ± 4.4 μg (range, 2.7-13.0 μg). The mean administered activity was 124 ± 20 MBq (range, 98-145 MBq). There were no adverse or clinically detectable pharmacologic effects in any of the subjects. No significant changes in vital signs, laboratory study results, or electrocardiography results were observed. Uptake of radioactivity was observed in the thyroid, salivary glands, liver, stomach wall, spleen, kidneys, ureters, and bladder. Time-activity curves indicated that the highest activity was in the bladder content, followed by the kidneys, small intestine, stomach, liver, spleen, thyroid, and gallbladder. The largest component of the residence times was the voided urine, followed by muscle, bladder, and liver. Using the mean residence time over all subjects as input to OLINDA/EXM, the effective dose was determined to be 0.0217 mSv/MBq; using residence times from single subjects gave an SD of 0.0020 mSv/MBq from the mean. The critical organ was the urinary bladder, with an absorbed dose of 0.18 mGy/MBq. Conclusion:18 F-FB-A20FMDV2 successfully passed toxicology criteria, showed no adverse effects in this first-in-humans study, and has an effective dose that enables multiple scans in a single subject., (© 2018 by the Society of Nuclear Medicine and Molecular Imaging.)- Published
- 2018
- Full Text
- View/download PDF
41. Safety, tolerability and pharmacokinetics of GSK3008348, a novel integrin αvβ6 inhibitor, in healthy participants.
- Author
-
Maden CH, Fairman D, Chalker M, Costa MJ, Fahy WA, Garman N, Lukey PT, Mant T, Parry S, Simpson JK, Slack RJ, Kendrick S, and Marshall RP
- Subjects
- Administration, Inhalation, Adult, Antigens, Neoplasm, Butyrates therapeutic use, Cross-Over Studies, Double-Blind Method, Female, Healthy Volunteers, Humans, Idiopathic Pulmonary Fibrosis drug therapy, Male, Middle Aged, Naphthyridines therapeutic use, Pyrazoles therapeutic use, Pyrrolidines therapeutic use, Butyrates pharmacokinetics, Butyrates pharmacology, Integrins antagonists & inhibitors, Naphthyridines pharmacokinetics, Naphthyridines pharmacology, Pyrazoles pharmacokinetics, Pyrazoles pharmacology, Pyrrolidines pharmacokinetics, Pyrrolidines pharmacology
- Abstract
Purpose: Inhaled drug delivery is an attractive route by which to deliver drugs to lungs of patients with idiopathic pulmonary fibrosis (IPF). GSK3008348 is a potent and selective small molecule being developed as the first inhaled inhibitor of the αvβ6 integrin for the treatment of IPF. The phase 1 first-time-in-human clinical trial (NCT02612051) presented here was designed to investigate the safety, tolerability and pharmacokinetic (PK) profile of single doses of GSK3008348 in healthy participants., Methods: Single ascending doses of GSK3008348 were administered to three cohorts of eight healthy participants in a randomised, double-blind, placebo-controlled, 4-period crossover design. Safety, tolerability and PK were assessed after single doses of 1-3000 mcg given by nebulisation., Results: A total of 29 participants were enrolled and received at least one dose of study treatment. There were no serious adverse events (AE) reported in any participant. No trends or clinically important differences were noted in the incidence or intensity of AEs or other safety assessments. Maximum plasma concentrations of GSK3008348 were generally attained within approximately 30 min after start of nebulisation, with geometric mean terminal elimination half-lives ranging from 7.95 to 10.2 h. Exposures, as measured by area under the plasma concentration-time curve (AUC), were dose proportional across all doses where estimates were possible (100-3000 mcg). Dose normalised geometric mean C
max increased with dose up to 3000 mcg. This supra proportionality was relatively modest, with a less than 3-fold increase over the range from 30 to 3000 mcg. The reason(s) for this observation are currently not known but may be due to slower absorption at the lowest doses. All exposures were within the exposure margins set by the non-clinical toxicity studies and so this is not expected to have any impact on safety., Conclusions: In summary, GSK3008348 was well tolerated at single doses up to 3000 mcg in healthy participants, and its PK profile was dose proportional at potentially clinically relevant doses (300-3000 mcg). These findings support further development of GSK3008348 as a novel inhaled treatment option for IPF.- Published
- 2018
- Full Text
- View/download PDF
42. An αv-RGD Integrin Inhibitor Toolbox: Drug Discovery Insight, Challenges and Opportunities.
- Author
-
Hatley RJD, Macdonald SJF, Slack RJ, Le J, Ludbrook SB, and Lukey PT
- Subjects
- Animals, Cell Adhesion drug effects, Humans, Integrin alphaV chemistry, Models, Molecular, Oligopeptides, Drug Discovery, Integrin alphaV metabolism
- Abstract
There is a requirement for efficacious and safe medicines to treat diseases with high unmet need. The resurgence in αv-RGD integrin inhibitor drug discovery is poised to contribute to this requirement. However, drug discovery in the αv integrin space is notoriously difficult due to the receptors being structurally very similar as well as the polar zwitterionic nature of the pharmacophore. This Review aims to guide drug discovery research in this field through an αv inhibitor toolbox, consisting of small molecules and antibodies. Small-molecule αv tool compounds with extended profiles in αvβ1, 3, 5, 6 and 8 cell adhesion assays, with key physicochemical properties, have been collated to assist in the selection of the right tool for the right experiment. This should also facilitate an understanding of partial selectivity profiles of compounds generated in different assays across research institutions. Prospects for further αv integrin research and the critical importance of target validation are discussed, where increased knowledge of the selectivity for individual RGD αv integrins is key. Insights into the design of small-molecule RGD chemotypes for topical or oral administration are provided and clinical findings on advanced molecules are examined., (© 2018 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2018
- Full Text
- View/download PDF
43. Identification of selective 8-(piperidin-4-yloxy)quinoline sulfone and sulfonamide histamine H 1 receptor antagonists for use in allergic rhinitis.
- Author
-
Procopiou PA, Ford AJ, Gore PM, Hancock AP, Hodgson ST, Holmes DS, Looker BE, Vile S, Clark KL, Saunders KA, Slack RJ, and Watts CJ
- Subjects
- Administration, Intranasal, Animals, Brain metabolism, Dogs, Guinea Pigs, Half-Life, Histamine H1 Antagonists pharmacokinetics, Histamine H1 Antagonists therapeutic use, Inhibitory Concentration 50, Quinolines pharmacokinetics, Quinolines therapeutic use, Rats, Receptors, Histamine H1 chemistry, Rhinitis, Allergic metabolism, Rhinitis, Allergic pathology, Structure-Activity Relationship, Sulfanilamide, Sulfanilamides pharmacokinetics, Sulfanilamides therapeutic use, Sulfonamides pharmacokinetics, Sulfonamides therapeutic use, Sulfones pharmacokinetics, Sulfones therapeutic use, Histamine H1 Antagonists chemistry, Quinolines chemistry, Receptors, Histamine H1 metabolism, Rhinitis, Allergic drug therapy, Sulfanilamides chemistry, Sulfonamides chemistry, Sulfones chemistry
- Abstract
A series of potent, selective and long-acting quinoline-based sulfonamide human H
1 histamine receptor antagonists, designed for once-daily intranasal administration for the treatment of rhinitis were developed. Sulfonamide 33b had a slightly lower affinity for the H1 receptor than azelastine, had low oral bioavailability in the rat and dog, and was turned over to five major metabolites. Furthermore, 33b had longer duration of action than azelastine in guinea pigs, lower rat brain-penetration, and did not cause time dependent inhibition of CYP2D6 or CYP3A4. The clinical dose in humans is expected to be low (approximately 0.5mg per day) based on the clinical dose used for azelastine and a comparison of efficacy data from animal models for 33b and azelastine., (Copyright © 2017. Published by Elsevier Ltd.)- Published
- 2017
- Full Text
- View/download PDF
44. Determining the True Selectivity Profile of αv Integrin Ligands Using Radioligand Binding: Applying an Old Solution to a New Problem.
- Author
-
Rowedder JE, Ludbrook SB, and Slack RJ
- Subjects
- Animals, CHO Cells, Cell Adhesion drug effects, Cricetinae, Cricetulus, Humans, K562 Cells, Kinetics, Ligands, Oligopeptides chemistry, Protein Binding drug effects, Small Molecule Libraries pharmacology, Tritium metabolism, Integrin alphaV metabolism, Radioligand Assay methods
- Abstract
The arginyl-glycinyl-aspartic acid (RGD) integrin subfamily contains five members that partner with the αv subunit: αvβ1, αvβ3, αvβ5, αvβ6, and αvβ8. Within the αv integrins, the epithelially restricted αvβ6 has been identified as playing a key role in the activation of transforming growth factor β that is hypothesized to be pivotal in the development of idiopathic pulmonary fibrosis (IPF). As part of a drug discovery program to identify a selective αvβ6 RGD mimetic for IPF, cell adhesion and radioligand binding assays were investigated to screen compounds to determine affinity and αv integrin selectivity. In this study, a pan-αv radioligand was characterized against all the αv integrins and used to determine accurate selectivity profiles for literature and novel RGD ligands, as well as enable an early readout on αvβ6 dissociation kinetics. It has been shown that while cell adhesion offers a high throughput and reliable format for ranking compounds, there are downsides to this format when comparing selectivity across αv integrins. By accurately defining the relationship between these assay formats, a medicinal chemistry effort has identified novel, high-affinity, and selective αvβ6 RGD mimetics with slow dissociation kinetics, with the potential to be developed into clinical candidates for IPF.
- Published
- 2017
- Full Text
- View/download PDF
45. Design of Phthalazinone Amide Histamine H 1 Receptor Antagonists for Use in Rhinitis.
- Author
-
Procopiou PA, Ford AJ, Gore PM, Looker BE, Hodgson ST, Holmes DS, Vile S, Clark KL, Saunders KA, Slack RJ, Rowedder JE, and Watts CJ
- Abstract
The synthesis of potent amide-containing phthalazinone H
1 histamine receptor antagonists is described. Three analogues 3e , 3g , and 9g were equipotent with azelastine and were longer-acting in vitro. Amide 3g had low oral bioavailability, low brain-penetration, high metabolic clearance, and long duration of action in vivo, and it was suitable for once-daily dosing intranasally, with a predicted dose for humans of approximately 0.5 mg per day.- Published
- 2017
- Full Text
- View/download PDF
46. The discovery of quinoline based single-ligand human H 1 and H 3 receptor antagonists.
- Author
-
Procopiou PA, Ancliff RA, Gore PM, Hancock AP, Hodgson ST, Holmes DS, Keeling SP, Looker BE, Parr NA, Rowedder JE, and Slack RJ
- Subjects
- Dose-Response Relationship, Drug, Histamine H1 Antagonists chemical synthesis, Histamine H1 Antagonists chemistry, Histamine H3 Antagonists chemical synthesis, Histamine H3 Antagonists chemistry, Humans, Ligands, Molecular Structure, Quinolines chemical synthesis, Quinolines chemistry, Structure-Activity Relationship, Drug Discovery, Histamine H1 Antagonists pharmacology, Histamine H3 Antagonists pharmacology, Quinolines pharmacology, Receptors, Histamine H1 metabolism, Receptors, Histamine H3 metabolism
- Abstract
A novel series of potent quinoline-based human H
1 and H3 bivalent histamine receptor antagonists, suitable for intranasal administration for the potential treatment of allergic rhinitis associated nasal congestion, were identified. Compound 18b had slightly lower H1 potency (pA2 8.8 vs 9.7 for the clinical goldstandard azelastine), and H3 potency (pKi 9.1vs 6.8 for azelastine), better selectivity over α1A , α1B and hERG, similar duration of action, making 18b a good back-up compound to our previous candidate, but with a more desirable profile., (Copyright © 2016 Elsevier Ltd. All rights reserved.)- Published
- 2016
- Full Text
- View/download PDF
47. Characterisation of a novel, high affinity and selective αvβ6 integrin RGD-mimetic radioligand.
- Author
-
Hall ER, Bibby LI, and Slack RJ
- Subjects
- Algorithms, Allosteric Site, Animals, Antigens, Neoplasm chemistry, Antigens, Neoplasm genetics, Antineoplastic Agents chemistry, Binding, Competitive, Biomimetics methods, Fibrinogen antagonists & inhibitors, Fibrinogen metabolism, HT29 Cells, Humans, Integrins antagonists & inhibitors, Integrins chemistry, Integrins genetics, Kinetics, Ligands, Mice, Naphthyridines chemistry, Naphthyridines pharmacology, Oligopeptides chemistry, Platelet Aggregation drug effects, Platelet Aggregation Inhibitors chemistry, Platelet Aggregation Inhibitors metabolism, Platelet Aggregation Inhibitors pharmacology, Radioligand Assay, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Solubility, Tritium, Antigens, Neoplasm metabolism, Antineoplastic Agents metabolism, Integrins metabolism, Naphthyridines metabolism, Oligopeptides metabolism
- Abstract
The alpha-v beta-6 (αvβ6) integrin has been identified as playing a key role in the activation of transforming growth factor-β (TGFβ) that is hypothesised to be pivotal in the development of cancer and fibrotic diseases. Therefore, the αvβ6 integrin is an attractive therapeutic target for these debilitating diseases and a drug discovery programme to identify small molecule αvβ6 selective arginyl-glycinyl-aspartic acid (RGD)-mimetics was initiated within GlaxoSmithKline. The primary aim of this study was to pharmacologically characterise the binding to αvβ6 of a novel clinical candidate, compound 1, using a radiolabelled form. Radioligand binding studies were completed with [(3)H]compound 1 against the human and mouse soluble protein forms of αvβ6 to determine accurate affinity estimates and binding kinetics. The selectivity of compound 1 for the RGD integrin family was also determined using saturation binding studies (αvβ1, αvβ3, αvβ5, αvβ8, α5β1 and α8β1 integrins) and fibrinogen-induced platelet aggregation (αIIbβ3 integrin). In addition, the relationship between divalent metal cation type and concentration and αvβ6 RGD site binding was also investigated. Compound 1 has been demonstrated to bind with extremely high affinity and selectivity for the αvβ6 integrin and has the potential as a clinical tool and therapeutic for investigating the role of αvβ6 in a range of disease states both pre-clinically and clinically. In addition, this is the first study that has successfully applied radioligand binding to the RGD integrin field to accurately determine the affinity and selectivity profile of a small molecule RGD-mimetic., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
48. 2,8-Diazaspiro[4.5]decan-8-yl)pyrimidin-4-amine potent CCR4 antagonists capable of inducing receptor endocytosis.
- Author
-
Shukla L, Ajram LA, Begg M, Evans B, Graves RH, Hodgson ST, Lynn SM, Miah AH, Percy JM, Procopiou PA, Richards SA, and Slack RJ
- Subjects
- Dose-Response Relationship, Drug, Humans, Molecular Structure, Pyrimidines chemical synthesis, Pyrimidines chemistry, Receptors, CCR4 metabolism, Spiro Compounds chemical synthesis, Spiro Compounds chemistry, Structure-Activity Relationship, Endocytosis drug effects, Pyrimidines pharmacology, Receptors, CCR4 antagonists & inhibitors, Spiro Compounds pharmacology
- Abstract
A number of potent 2,8-diazaspiro[4.5]decan-8-yl)pyrimidin-4-amine CCR4 antagonists binding to the extracellular allosteric site were synthesised. (R)-N-(2,4-Dichlorobenzyl)-2-(2-(pyrrolidin-2-ylmethyl)-2,8-diazaspiro[4.5]decan-8-yl)pyrimidin-4-amine (R)-(18a) has high affinity in both the [(125)I]-TARC binding assay with a pKi of 8.8, and the [(35)S]-GTPγS functional assay with a pIC50 of 8.1, and high activity in the human whole blood actin polymerisation assay (pA2 = 6.7). The most potent antagonists were also investigated for their ability to induce endocytosis of CCR4 and were found to internalise about 60% of the cell surface receptors, a property which is not commonly shared by small molecule antagonists of chemokine receptors., (Copyright © 2016 Elsevier Masson SAS. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
49. Pharmacological Characterization of the αvβ6 Integrin Binding and Internalization Kinetics of the Foot-and-Mouth Disease Virus Derived Peptide A20FMDV2.
- Author
-
Slack RJ, Hafeji M, Rogers R, Ludbrook SB, Marshall JF, Flint DJ, Pyne S, and Denyer JC
- Subjects
- Binding Sites, Cell Line, Humans, Kinetics, Ligands, Protein Binding, Radioligand Assay, Antigens, Neoplasm metabolism, Foot-and-Mouth Disease Virus, Integrins metabolism, Peptides metabolism
- Abstract
A20FMDV2 is a peptide derived from the foot-and-mouth disease virus with a high affinity and selectivity for the alpha-v beta-6 (αvβ6) arginyl-glycinyl-aspartic acid (RGD)-binding integrin. It has been shown to be an informative tool ligand in pre-clinical imaging studies for selective labelling of the αvβ6 integrin in a number of disease models. In a radioligand binding assay using a radiolabelled form of the peptide ([3H]A20FMDV2), its high affinity (K(D): 0.22 nmol/l) and selectivity (at least 85-fold) for αvβ6 over the other members of the RGD integrin family was confirmed. [3H]A20FMDV2 αvβ6 binding could be fully reversed only in the presence of EDTA, whereas a partial reversal was observed in the presence of excess concentrations of an RGD-mimetic small molecule (SC-68448) or unlabelled A20FMDV2. Using flow cytometry on bronchial epithelial cells, the ligand-induced internalization of αvβ6 by A20FMDV2 and latency-associated peptide-1 was shown to be fast (t(1/2): 1.5 and 3.1 min, respectively), concentration-dependent (EC50: values 1.1 and 3.6 nmol/l, respectively) and was followed by a moderately slow return of integrin to the surface. The results of the radioligand binding studies suggest that the binding of A20FMDV2 to the RGD-binding site on αvβ6 is required to maintain its engagement with the hypothesised A20FMDV2 synergy site on the integrin. In addition, there is evidence from flow cytometric studies that the RGD-ligand engagement of αvβ6 post-internalization plays a role in delaying recycling of the integrin to the cell surface. This mechanism may act as a homeostatic control of membrane αvβ6 following RGD ligand engagement., (© 2016 S. Karger AG, Basel.)
- Published
- 2016
- Full Text
- View/download PDF
50. Antagonism of human CC-chemokine receptor 4 can be achieved through three distinct binding sites on the receptor.
- Author
-
Slack RJ, Russell LJ, Barton NP, Weston C, Nalesso G, Thompson SA, Allen M, Chen YH, Barnes A, Hodgson ST, and Hall DA
- Abstract
Chemokine receptor antagonists appear to access two distinct binding sites on different members of this receptor family. One class of CCR4 antagonists has been suggested to bind to a site accessible from the cytoplasm while a second class did not bind to this site. In this report, we demonstrate that antagonists representing a variety of structural classes bind to two distinct allosteric sites on CCR4. The effects of pairs of low-molecular weight and/or chemokine CCR4 antagonists were evaluated on CCL17- and CCL22-induced responses of human CCR4(+) T cells. This provided an initial grouping of the antagonists into sets which appeared to bind to distinct binding sites. Binding studies were then performed with radioligands from each set to confirm these groupings. Some novel receptor theory was developed to allow the interpretation of the effects of the antagonist combinations. The theory indicates that, generally, the concentration-ratio of a pair of competing allosteric modulators is maximally the sum of their individual effects while that of two modulators acting at different sites is likely to be greater than their sum. The low-molecular weight antagonists could be grouped into two sets on the basis of the functional and binding experiments. The antagonistic chemokines formed a third set whose behaviour was consistent with that of simple competitive antagonists. These studies indicate that there are two allosteric regulatory sites on CCR4.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.