33 results on '"Tjeerd Barf"'
Search Results
2. Identification and Characterization of ACP-5862, the Major Circulating Active Metabolite of Acalabrutinib: Both Are Potent and Selective Covalent Bruton Tyrosine Kinase Inhibitors
- Author
-
Terry, Podoll, Paul G, Pearson, Allard, Kaptein, Jerry, Evarts, Gerjan, de Bruin, Maaike, Emmelot-van Hoek, Anouk, de Jong, Bart, van Lith, Hao, Sun, Stephen, Byard, Adrian, Fretland, Niels, Hoogenboom, Tjeerd, Barf, and J Greg, Slatter
- Subjects
Pharmacology ,Molecular Medicine - Abstract
Acalabrutinib is a covalent Bruton tyrosine kinase (BTK) inhibitor approved for relapsed/refractory mantle cell lymphoma and chronic lymphocytic leukemia/small lymphocytic lymphoma. A major metabolite of acalabrutinib (M27, ACP-5862) was observed in human plasma circulation. Subsequently, the metabolite was purified from an in vitro biosynthetic reaction and shown by nuclear magnetic resonance spectroscopy to be a pyrrolidine ring-opened ketone/amide. Synthesis confirmed its structure, and covalent inhibition of wild-type BTK was observed in a biochemical kinase assay. A twofold lower potency than acalabrutinib was observed but with similar high kinase selectivity. Like acalabrutinib, ACP-5862 was the most selective toward BTK relative to ibrutinib and zanubrutinib. Because of the potency, ACP-5862 covalent binding properties, and potential contribution to clinical efficacy of acalabrutinib, factors influencing acalabrutinib clearance and ACP-5862 formation and clearance were assessed. rCYP (recombinant cytochrome P450) reaction phenotyping indicated that CYP3A4 was responsible for ACP-5862 formation and metabolism. ACP-5862 formation K
- Published
- 2022
3. Discovery and optimization of covalent EGFR T790M/L858R mutant inhibitors
- Author
-
Maaike Emmelot, Dennis Demont, Edwin de Zwart, Allard Kaptein, Bas van de Kar, Saskia Verkaik, Tjeerd Barf, and Niels Hoogenboom
- Subjects
Models, Molecular ,Clinical Biochemistry ,Mutant ,Pharmaceutical Science ,medicine.disease_cause ,Afatinib ,Biochemistry ,chemistry.chemical_compound ,T790M ,Structure-Activity Relationship ,Drug Discovery ,medicine ,Humans ,Epidermal growth factor receptor ,Molecular Biology ,Protein Kinase Inhibitors ,Mutation ,Acrylamides ,Aniline Compounds ,biology ,Dose-Response Relationship, Drug ,Molecular Structure ,Organic Chemistry ,Wild type ,Rash ,respiratory tract diseases ,ErbB Receptors ,chemistry ,Covalent bond ,Cancer research ,biology.protein ,Molecular Medicine ,medicine.symptom ,Lead compound - Abstract
Epidermal growth factor receptor (EGFR) inhibitors have clinical utility in the treatment of non-small cell lung cancer (NSCLC) patients. Despite encouraging clinical efficacy with these agents, many patients develop resistance due to sensitizing (or activating) mutations ultimately leading to disease progression. In the majority of the cases, this resistance is due to the T790M mutation and frequently coexisting L858R. In addition, EGFR wild type receptor inhibition can lead to on target related dose limiting toxicities such as rash and diarrhea. We describe herein the identification of a mutant selective lead compound 12, an irreversible covalent inhibitor of EGFR T790M/L858R resistance mutations with selectivity over the wild type form. Significant tumor growth inhibition in preclinical models was observed with this lead.
- Published
- 2021
4. Abstract 4049: Preclinical pharmacology profile of GTX-0196: A novel, potent and highly selective dihydroorotate dehydrogenase (DHODH) inhibitor for the treatment of hematologic malignancies
- Author
-
Allard Kaptein, Sonia Lain, Diede Brunen, Edward van Wezel, and Tjeerd Barf
- Subjects
Cancer Research ,Oncology - Abstract
Dihydroorotate dehydrogenase (DHODH) is a key enzyme in the de novo pyrimidine synthesis [Löffler et al., 1997]. Initial clinical studies with the DHODH inhibitor brequinar in solid tumors showed limited efficacy due to dose limiting adverse effects [Peters et al, 2018]. Recently, there is renewed interest in the use of more potent and selective DHODH inhibitors for the treatment of myeloid and lymphoid malignancies [Sykes et al, 2016]. Here we report the identification of the novel potent and highly selective DHODH inhibitor GTX-0196, with excellent physicochemical properties. GTX-0196 exhibited a biochemical IC50 of 3.7 nM in a DHODH biochemical assay and an antiproliferative IC50 of 1.1 nM in the MOLM-13 cell line, with brequinar showing IC50 values of 6.9 and 50 nM, respectively. The activity in the MOLM-13 proliferation assay was uridine-dependent, indicating absence of general toxicity of the compound and confirming DHODH-mediated efficacy. Profiling of GTX-0196 in the BioPrint panel (Eurofins/CEREP, at 10 µM), showed 2000-fold selectivity versus other targets tested). Furthermore, no inhibition was observed in a human CYP panel (1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1 and 3A4, at 30 µM) and in a panel of transporters (at 10 µM). High metabolic stability in human liver microsomes and hepatocytes was observed with t½ > 6h and 10h, respectively. Compared to an earlier lead compound, GTX-0196 demonstrated enhanced potency and metabolic stability (particularly in preclinical species). Potent and efficacious inhibition of proliferation through DHODH was confirmed in a large number of solid tumor and hematologic malignancy cell lines, illustrating the broad potential use of this chemical series. Furthermore, tumor growth inhibition was observed in the MOLM-13 xenograft mouse model. Due to its favorable properties, GTX-0196 has progressed towards IND-enabling studies. In parallel, we are exploring mono and combination therapy opportunities in hematological malignancies as well as combination therapy opportunities in solid tumors to investigate the full therapeutic potential in oncology for this optimized DHODH inhibitor. Löffler et al. Mol. Cell. Biochem. 1997Peters GJ et al, Nucleosides, Nucleotides and Nucleic Acids 2018Sykes DB et al. Cell 2016 Citation Format: Allard Kaptein, Sonia Lain, Diede Brunen, Edward van Wezel, Tjeerd Barf. Preclinical pharmacology profile of GTX-0196: A novel, potent and highly selective dihydroorotate dehydrogenase (DHODH) inhibitor for the treatment of hematologic malignancies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 4049.
- Published
- 2022
5. Chemical genetics strategy to profile kinase target engagement reveals role of FES in neutrophil phagocytosis
- Author
-
Tim van den Hooven, Joost A. P. M. Wijnakker, Tjeerd Barf, Herman S. Overkleeft, Hans den Dulk, Rob Ruijtenbeek, Tom van der Wel, Eelke B. Lenselink, Allard Kaptein, Gerard J. P. van Westen, Bogdan I. Florea, Nienke M. Prins, Mario van der Stelt, and Riet Hilhorst
- Subjects
0301 basic medicine ,Neutrophils ,Science ,Cellular differentiation ,General Physics and Astronomy ,Kinases ,Article ,Chemical genetics ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Phagocytosis ,Fluorescence Resonance Energy Transfer ,Humans ,Syk Kinase ,CRISPR ,lcsh:Science ,Sensors and probes ,Fluorescent Dyes ,Gene Editing ,Multidisciplinary ,Drug discovery ,Chemistry ,Kinase ,Cas9 ,Macrophages ,Cell Differentiation ,General Chemistry ,Protein-Tyrosine Kinases ,Cell biology ,030104 developmental biology ,Proto-Oncogene Proteins c-fes ,030220 oncology & carcinogenesis ,Mutation ,lcsh:Q ,ATP-Binding Cassette Transporters ,CRISPR-Cas Systems ,Signal transduction ,Chemical tools ,Tyrosine kinase ,Signal Transduction - Abstract
Chemical tools to monitor drug-target engagement of endogenously expressed protein kinases are highly desirable for preclinical target validation in drug discovery. Here, we describe a chemical genetics strategy to selectively study target engagement of endogenous kinases. By substituting a serine residue into cysteine at the DFG-1 position in the ATP-binding pocket, we sensitize the non-receptor tyrosine kinase FES towards covalent labeling by a complementary fluorescent chemical probe. This mutation is introduced in the endogenous FES gene of HL-60 cells using CRISPR/Cas9 gene editing. Leveraging the temporal and acute control offered by our strategy, we show that FES activity is dispensable for differentiation of HL-60 cells towards macrophages. Instead, FES plays a key role in neutrophil phagocytosis via SYK kinase activation. This chemical genetics strategy holds promise as a target validation method for kinases., Chemical tools to monitor drug-target engagement of endogenous enzymes are essential for preclinical target validation. Here, the authors present a chemical genetics strategy to study target engagement of endogenous kinases, achieving specific labeling and inactivation of FES kinase to provide insights into FES’ role in neutrophil phagocytosis.
- Published
- 2020
6. Discovery of quinoline-based irreversible BTK inhibitors
- Author
-
Tjeerd Barf, Bart Van Lith, Todd Covey, Dennis Demont, Niels Hoogenboom, Bas van de Kar, Saskia Verkaik, Allard Kaptein, Maaike van Hoek, Gerjan de Bruin, Michael Gulrajani, and Edwin de Zwart
- Subjects
Models, Molecular ,Clinical Biochemistry ,Pharmaceutical Science ,01 natural sciences ,Biochemistry ,chemistry.chemical_compound ,Structure-Activity Relationship ,immune system diseases ,In vivo ,hemic and lymphatic diseases ,Drug Discovery ,Quinazoline ,Agammaglobulinaemia Tyrosine Kinase ,Bruton's tyrosine kinase ,Humans ,Molecular Biology ,Protein Kinase Inhibitors ,biology ,Dose-Response Relationship, Drug ,Molecular Structure ,010405 organic chemistry ,Btk inhibitors ,Kinase ,Organic Chemistry ,Quinoline ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,chemistry ,biology.protein ,Cancer research ,Quinolines ,Molecular Medicine - Abstract
Bruton tyrosine kinase (BTK) is an important target in oncology and (auto)immunity. Various BTK inhibitors have been approved or are currently in clinical development. A novel BTK inhibitor series was developed starting with a quinazoline core. Moving from a quinazoline to a quinoline core provided a handle for selectivity for BTK over EGFR and resulted in the identification of potent and selective BTK inhibitors with good potency in human whole blood assay. Furthermore, proof of concept of this series for BTK inhibition was shown in an in vivo mouse model using one of the compounds identified.
- Published
- 2020
7. Chemical genetics strategy to profile kinase target engagement reveals role of FES in neutrophil phagocytosis via SYK activation
- Author
-
Nienke M. Prins, Rob Ruijtenbeek, Bogdan I. Florea, Joost A. P. M. Wijnakker, Mario van der Stelt, Hans den Dulk, Eelke B. Lenselink, Allard Kaptein, Tom van der Wel, Riet Hilhorst, Gerard J. P. van Westen, Tim van den Hooven, Tjeerd Barf, and Herman S. Overkleeft
- Subjects
Serine ,Immune system ,Chemistry ,Kinase ,Regulator ,Syk ,Chemical genetics ,Tyrosine kinase ,Small molecule ,Cell biology - Abstract
Chemical tools and methods that report on target engagement of endogenously expressed protein kinases by small molecules in human cells are highly desirable. Here, we describe a chemical genetics strategy that allows the study of non-receptor tyrosine kinase FES, a promising therapeutic target for cancer and immune disorders. Precise gene editing was used in combination with a rationally designed, complementary fluorescent probe to visualize endogenous FES kinase in HL-60 cells. We replaced a single oxygen atom by a sulphur in a serine residue at the DFG-1 position of the ATP-binding pocket in an endogenously expressed kinase, thereby sensitizing the engineered protein towards covalent labeling and inactivation by a fluorescent probe. The temporal control offered by this strategy allows acute inactivation of FES activity both during myeloid differentiation and in terminally differentiated neutrophils. Our results show that FES activity is dispensable for differentiation of HL-60 cells towards macrophages. Instead, FES plays a key role in neutrophil phagocytosis by activation of SYK kinase, a central regulator of immune function in neutrophils. This strategy holds promise as a target validation method for kinases.
- Published
- 2019
- Full Text
- View/download PDF
8. CHAPTER 4. Covalent Inhibition of Kinases
- Author
-
Gerjan de Bruin and Tjeerd Barf
- Subjects
Enzyme inhibition ,chemistry.chemical_compound ,Biochemistry ,Kinase ,Covalent bond ,Drug discovery ,Chemistry ,Afatinib ,Ibrutinib ,medicine ,Acalabrutinib ,Osimertinib ,medicine.drug - Abstract
In recent years, the field of covalent kinase drug discovery has been rapidly evolving. Several covalent kinase inhibitors (such as afatinib, osimertinib, ibrutinib and acalabrutinib) have obtained market approval or are currently in early and advanced clinical studies. Both covalent irreversible and covalent reversible kinase inhibitors have been discovered, providing an opportunity to target specific kinases, mutations and or pharmacokinetic (PK)/pharmacodynamic (PD) profiles. This chapter will give an overview of the state-of-the-art of covalent kinase drug discovery. First, the concept of covalent enzyme inhibition will be introduced and a comparison to non-covalent inhibitors will be made, highlighting possible advantages and disadvantages of such mechanisms of action (MOA). The importance of selectivity, drug-target residence times and inhibitor kinetics will also be discussed. Finally, a literature overview of irreversible and reversible covalent kinase inhibitors that are currently on the market or in the clinical testing phase will be presented.
- Published
- 2018
9. Direct and two-step bioorthogonal probes for Bruton's tyrosine kinase based on ibrutinib: a comparative study
- Author
-
Nora Liu, Tjeerd Barf, Sascha Hoogendoorn, Gijsbert A. van der Marel, Bas van de Kar, Christoph Driessen, Dmitri V. Filippov, Allard Kaptein, Mario van der Stelt, and Herman S. Overkleeft
- Subjects
Biochemistry ,chemistry.chemical_compound ,Piperidines ,immune system diseases ,hemic and lymphatic diseases ,Agammaglobulinaemia Tyrosine Kinase ,medicine ,Humans ,Bruton's tyrosine kinase ,Physical and Theoretical Chemistry ,Mode of action ,Protein Kinase Inhibitors ,Netherlands ,biology ,Adenine ,Organic Chemistry ,Macroglobulinemia ,Protein-Tyrosine Kinases ,medicine.disease ,3. Good health ,Pyrimidines ,chemistry ,Hematologic Neoplasms ,Molecular Probes ,Ibrutinib ,Biotinylation ,Cancer research ,biology.protein ,Pyrazoles ,Mantle cell lymphoma ,Bioorthogonal chemistry ,Tyrosine kinase - Abstract
Ibrutinib is a covalent and irreversible inhibitor of Bruton's tyrosine kinase (BTK) and has been approved for the treatment of haematological malignancies, such as chronic lymphocytic leukaemia, mantle cell lymphoma and Waldenström's macroglobulinemia. The covalent and irreversible nature of its molecular mode of action allows identification and monitoring of its target in an activity-based protein profiling (ABPP) setting. Fluorescent and biotinylated ibrutinib derivatives have appeared in the literature in recent years to monitor BTK in vitro and in situ. The work described here complements this existing methodology and pertains a comparative study on the efficacy of direct and two-step bioorthogonal ABPP of BTK.
- Published
- 2015
10. Acalabrutinib (ACP-196): A Covalent Bruton Tyrosine Kinase Inhibitor with a Differentiated Selectivity and In Vivo Potency Profile
- Author
-
Dennis Demont, Roger G. Ulrich, Tjeerd Barf, Raquel Izumi, Allard Kaptein, Bart Van Lith, Edwin de Zwart, Michael Gulrajani, Todd Covey, Saskia Verkaik, Paul G. Pearson, Diana Mittag, Bas van de Kar, Fanny Krantz, and Maaike van Hoek
- Subjects
0301 basic medicine ,Pharmacology ,Peripheral blood mononuclear cell ,03 medical and health sciences ,chemistry.chemical_compound ,Jurkat Cells ,Mice ,0302 clinical medicine ,In vivo ,Agammaglobulinaemia Tyrosine Kinase ,Bruton's tyrosine kinase ,Animals ,Humans ,Spebrutinib ,Epidermal growth factor receptor ,Protein Kinase Inhibitors ,Mice, Inbred BALB C ,biology ,Dose-Response Relationship, Drug ,Kinase ,Protein-Tyrosine Kinases ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Ibrutinib ,Pyrazines ,Benzamides ,biology.protein ,Leukocytes, Mononuclear ,Molecular Medicine ,Acalabrutinib - Abstract
Several small-molecule Bruton tyrosine kinase (BTK) inhibitors are in development for B cell malignancies and autoimmune disorders, each characterized by distinct potency and selectivity patterns. Herein we describe the pharmacologic characterization of BTK inhibitor acalabrutinib [compound 1, ACP-196 (4-[8-amino-3-[(2S)-1-but-2-ynoylpyrrolidin-2-yl]imidazo[1,5-a]pyrazin-1-yl]-N-(2-pyridyl)benzamide)]. Acalabrutinib possesses a reactive butynamide group that binds covalently to Cys481 in BTK. Relative to the other BTK inhibitors described here, the reduced intrinsic reactivity of acalabrutinib helps to limit inhibition of off-target kinases having cysteine-mediated covalent binding potential. Acalabrutinib demonstrated higher biochemical and cellular selectivity than ibrutinib and spebrutinib (compounds 2 and 3, respectively). Importantly, off-target kinases, such as epidermal growth factor receptor (EGFR) and interleukin 2-inducible T cell kinase (ITK), were not inhibited. Determination of the inhibitory potential of anti-immunoglobulin M-induced CD69 expression in human peripheral blood mononuclear cells and whole blood demonstrated that acalabrutinib is a potent functional BTK inhibitor. In vivo evaluation in mice revealed that acalabrutinib is more potent than ibrutinib and spebrutinib. Preclinical and clinical studies showed that the level and duration of BTK occupancy correlates with in vivo efficacy. Evaluation of the pharmacokinetic properties of acalabrutinib in healthy adult volunteers demonstrated rapid absorption and fast elimination. In these healthy individuals, a single oral dose of 100 mg showed approximately 99% median target coverage at 3 and 12 hours and around 90% at 24 hours in peripheral B cells. In conclusion, acalabrutinib is a BTK inhibitor with key pharmacologic differentiators versus ibrutinib and spebrutinib and is currently being evaluated in clinical trials.
- Published
- 2017
11. Abstract 2194: Preclinical pharmacological profiling of ACP-5862, the major metabolite of the covalent BTK inhibitor acalabrutinib, displays intrinsic BTK inhibitory activity
- Author
-
Bart Van Lith, Niels Hoogenboom, Gerjan de Bruin, J. Greg Slatter, Terry Podoll, Anouk de Jong, Maaike Emmelot-van Hoek, Tjeerd Barf, Joseph A. Ware, and Allard Kaptein
- Subjects
Cancer Research ,biology ,Kinase ,Metabolite ,Peripheral blood mononuclear cell ,Molecular biology ,chemistry.chemical_compound ,Oncology ,chemistry ,In vivo ,biology.protein ,Bruton's tyrosine kinase ,Acalabrutinib ,IC50 ,ADME - Abstract
Acalabrutinib is a potent and highly selective, oral covalent inhibitor of Bruton tyrosine kinase (BTK) that received accelerated approval for relapsed/refractory mantle cell lymphoma by the United States Food and Drug Administration in October 2017. ADME studies in humans, rat and dog, revealed extensive metabolism of acalabrutinib. The major circulating metabolite (M27, ACP-5862) was produced by CYP3A oxidation, resulting in a pyrrolidine ring-opened metabolite, with the butynamide electrophile still present. We investigated the on-target BTK inhibition and kinase selectivity profile of ACP-5862 using the same biochemical and cellular assays previously employed to profile acalabrutinib.1 The apparent BTK IC50 was determined over time using the LanthaScreen assay. Results indicate that ACP-5862, like acalabrutinib, is a covalent inhibitor of BTK. Binding kinetics derived from IC50 over time data, indicated that acalabrutinib and ACP-5862 have similar affinity (KI), however the BTK inactivation rate (kinact) for ACP-5862 was half that relative to acalabrutinib. This indicated a covalent BTK inhibition potency for ACP-5862 that was 2-fold lower than acalabrutinib. The overall kinome inhibition profile at 1 µM using KINOMEscan (Eurofins DiscoverX), as well as IC50 determinations on closely related kinases with a Cys in the same position as Cys481 in BTK (ITK, TXK, TEC, BMX, EGFR, ERBB2, ERBB4, BLK, JAK3), both revealed that the kinase selectivity profiles of acalabrutinib and ACP-5862 were similar. On-target inhibition in B cells was investigated measuring inhibition of B-cell antigen receptor-mediated activation of CD69 cell surface expression on human peripheral B cells using human peripheral blood mononuclear cells and human whole blood (hWB). The EC50 for anti-IgD-induced CD69 expression in hWB was 64 ± 6 nM for ACP-5862, compared to 9.2 ± 4.4 nM for acalabrutinib. The hWB EC90, representing near complete inhibition of BTK by acalabrutinib and ACP-5862 was observed at 72 ± 20 nM and 544 ± 376 nM, respectively. In conclusion, the results indicate that ACP-5862, the major metabolite of acalabrutinib, has intrinsic BTK inhibitory activity and a similar kinase selectivity profile as acalabrutinib. The contribution of ACP-5862 to on-target covalent inhibition of BTK in humans is unclear at present, but is most likely limited, since the EC90 in hWB for ACP-5862 approximated observed plasma Cmax in humans dosed with 100 mg acalabrutinib. The relative contribution of acalabrutinib and ACP-5862 to BTK pharmacodynamics is under further investigation. Terry Podoll and J. Greg Slatter are former Acerta Pharma employees. Reference 1. Barf T, Covey T, Izumi R, et al. Acalabrutinib (ACP-196): A covalent Bruton tyrosine kinase inhibitor with a differentiated selectivity and in vivo potency profile. J Pharmacol Exp Ther. 2017;363(2):240-252. Citation Format: Allard Kaptein, Terry Podoll, Gerjan de Bruin, Maaike Emmelot-van Hoek, Anouk de Jong, Bart van Lith, Niels Hoogenboom, Tjeerd Barf, Joseph Ware, J. Greg Slatter. Preclinical pharmacological profiling of ACP-5862, the major metabolite of the covalent BTK inhibitor acalabrutinib, displays intrinsic BTK inhibitory activity [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 2194.
- Published
- 2019
12. Irreversible Protein Kinase Inhibitors: Balancing the Benefits and Risks
- Author
-
Tjeerd Barf and Allard Kaptein
- Subjects
Risk ,Time Factors ,business.industry ,Chemistry ,Computational biology ,Cell Line ,Structure-Activity Relationship ,Text mining ,Drug Discovery ,Animals ,Humans ,Molecular Medicine ,Structure–activity relationship ,business ,Protein kinase A ,Protein Kinase Inhibitors ,Protein Kinases - Published
- 2012
13. Potency and Selectivity of BTK Inhibitors in Clinical Development for B-Cell Malignancies
- Author
-
Diana Mittag, Bas van de Kar, Tjeerd Barf, Dennis Demont, Todd Covey, Anouk de Jong, Gerjan de Bruin, Maaike Emmelot-van Hoek, Allard Kaptein, and Michael Gulrajani
- Subjects
0301 basic medicine ,Cancer Research ,biology ,Kinase ,Immunology ,Cell Biology ,Hematology ,Pharmacology ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Ibrutinib ,biology.protein ,Acalabrutinib ,Bruton's tyrosine kinase ,Spebrutinib ,Kinome ,IL-2 receptor ,IC50 - Abstract
Background: Bruton tyrosine kinase (BTK) is a validated target for B-cell malignancies. The BTK inhibitor ibrutinib was approved in chronic lymphocytic leukemia, mantle cell lymphoma (MCL), and Waldenstrom macroglobulinemia. Acalabrutinib is a potent, highly selective, covalent BTK inhibitor with minimal off-target activity; it received accelerated FDA approval in October 2017 for the treatment of patients with MCL having ≥1 prior therapy. In addition to the approved covalent BTK inhibitors ibrutinib and acalabrutinib, clinical data in B-cell malignancies are available for spebrutinib (CC-292), tirabrutinib (ONO/GS-4059) and zanubrutinib (BGB-3111). We performed biochemical and cellular profiling of these 5 BTK inhibitors, investigating potency and selectivity. Methods: Two biochemical kinase assays assessed BTK inhibitor potency, with IC50 determination at a fixed time point (IMAP; Molecular Devices) or over time (LanthaScreen; Invitrogen); the latter was used to calculate binding kinetics. Kinome profiling was performed at a single dose (1 µM) using KINOMEscan (Eurofins DiscoverX). IC50 determinations were made using kinases with a Cys in the same position as the Cys481 residue in BTK, using assays developed in house (using IMAP and LanthaScreen) or at Thermo Fisher Scientific (Z'-LYTE) with IC50 determination at a fixed time point. On-target inhibition of BTK in cellular assays was evaluated using B-cell receptor-mediated activation of CD69 expression on peripheral B cells using human peripheral blood mononuclear cells (hPBMCs) or human whole blood (hWB). Off-target inhibition of epidermal growth factor receptor (EGFR) was evaluated in a cellular assay examining the effect of EGF-induced EGFR phosphorylation in A431 cells. Off-target inhibition of ITK and/or TXK was evaluated using T-cell receptor (TCR)-mediated activation of interleukin-2 expression in Jurkat T cells and CD25 cell surface expression using primary human peripheral T cells. Results: Based on biochemical binding kinetics, ibrutinib and zanubrutinib were the most potent BTK inhibitors (Table 1), followed by spebrutinib; acalabrutinib and tirabrutinib had comparable potency. Differences in potency were largely driven by differences in inactivation rates. However, differences in biochemical potency were lost (in part) in cellular assays using hPBMCs or hWB. EC50 values in hWB were Differences in overall kinase selectivity were observed among the BTK inhibitors (Figure 1; KINOMEscan). Acalabrutinib had the lowest hit rate; 1.5% of human wild-type kinases were inhibited >65% at 1 µM (excluding BTK). Kinase hit rate was also low for tirabrutinb (2.3%), whereas ibrutinib (9.4%), zanubrutinib (4.3%), and spebrutinib (8.3%) had higher hit rates. Acalabrutinib had a high selectivity for BTK over kinases with a Cys in the same position as the Cys481 residue in BTK (Table 2). Similar results were observed for tirabrutinib, whereas ibrutinib, spebrutinib and zanubrutinib were less selective in this panel of kinases with potential for off-target covalent binding by BTK inhibitors (Table 2). Acalabrutinib also had a higher selectivity for BTK over Src-family kinases than the other BTK inhibitors tested. Acalabrutinib and tirabrutinib had EC50 values >10 µM in the cellular assay of off-target inhibition of EGFR; ibrutinib, zanubrutinib, and spebrutinib had EC50 values of 0.07, 0.39, and 4.7 µM, respectively. No off-target inhibition was observed for acalabrutinib or tirabrutinib on TCR-mediated activation of T cells up to 10 µM. Ibrutinib, zanubrutinib, and spebrutinib had EC50 values Conclusion: BTK inhibitors in clinical development for B-cell malignancies had differing potency in biochemical assays, but these differences were lost (in part) in cellular assays, particularly in hWB. Among the BTK inhibitors tested, the greatest differentiation was observed in kinase selectivity profiles; acalabrutinib and tirabrutinib had the highest kinase selectivity. Disclosures Kaptein: Covaluation Pharma BV: Employment, Equity Ownership; Acerta Pharma BV: Consultancy, Equity Ownership; Apo-T BV: Consultancy. de Bruin:Acerta Pharma: Employment. Emmelot-van Hoek:Acerta Pharma: Employment. van de Kar:Acerta Pharma: Employment. de Jong:Acerta Pharma: Employment. Gulrajani:Acerta Pharma: Employment, Equity Ownership. Demont:Acerta Pharma: Employment. Covey:AstraZeneca: Equity Ownership; Acerta Pharma: Employment. Mittag:Acerta Pharma: Employment, Equity Ownership. Barf:Covaluation Holding BV: Employment, Equity Ownership; Acerta Pharma BV: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees.
- Published
- 2018
14. Characterization of Irreversible Kinase Inhibitors by Directly Detecting Covalent Bond Formation: A Tool for Dissecting Kinase Drug Resistance
- Author
-
Christian Grütter, Daniel Rauh, Tjeerd Barf, Willem A. L. van Otterlo, Jeffrey R. Simard, Matthias Rabiller, Vijaykumar Pawar, Hans C.A. Raaijmakers, Haridas B. Rode, and Sabine Klüter
- Subjects
Models, Molecular ,Steric effects ,Mutant ,Crystallography, X-Ray ,medicine.disease_cause ,Biochemistry ,Neoplasms ,medicine ,Animals ,Humans ,Molecule ,Kinase activity ,Protein Kinase Inhibitors ,Molecular Biology ,Mutation ,Molecular Structure ,Chemistry ,Kinase ,Organic Chemistry ,ErbB Receptors ,Spectrometry, Fluorescence ,src-Family Kinases ,Structural biology ,Drug Resistance, Neoplasm ,Covalent bond ,Molecular Medicine ,Chickens ,Protein Kinases - Abstract
Targeting protein kinases in cancer therapy with irreversible small-molecule inhibitors is moving to the forefront of kinase-inhibitor research and is thought to be an effective means of overcoming mutation-associated drug resistance in epidermal growth factor receptor kinase (EGFR). We generated a detection technique that allows direct measurements of covalent bond formation without relying on kinase activity, thereby allowing the straightforward investigation of the influence of steric clashes on covalent inhibitors in different resistant kinase mutants. The obtained results are discussed together with structural biology and biochemical studies of catalytic activity in both wild-type and gatekeeper mutated kinase variants to draw conclusions about the impact of steric hindrance and increased catalytic activity in drug-resistant kinase variants.
- Published
- 2010
15. Intervention of Hepatic Glucose Production. Small Molecule Regulators of Potential Targets for Type 2 Diabetes Therapy
- Author
-
Tjeerd Barf
- Subjects
medicine.medical_specialty ,Molecular Conformation ,Dehydrogenase ,Type 2 diabetes ,Models, Biological ,chemistry.chemical_compound ,Glycogen phosphorylase ,Receptors, Glucocorticoid ,Glucocorticoid receptor ,Internal medicine ,Drug Discovery ,Receptors, Glucagon ,medicine ,Animals ,Humans ,Hypoglycemic Agents ,Glycogen synthase ,Pharmacology ,biology ,Drug discovery ,Gluconeogenesis ,Fructose ,General Medicine ,medicine.disease ,Enzymes ,Glucose ,Endocrinology ,Diabetes Mellitus, Type 2 ,Liver ,chemistry ,biology.protein ,Glucagon receptor ,Glycogen - Abstract
Excessive hepatic glucose production is thought to be a major contributor to the type 2 diabetic state. Drug discovery efforts have yielded small synthetic inhibitors for gluconeogenic and glycogenic regulators of this pathway. The most advanced targets are outlined in this mini-review and include: the glucocorticoid receptor, 11 beta-hydroxysteroid dehydrogenase type 1, fructose 1,6-bisphosphatase, the glucagon receptor, glycogen phosphorylase, glycogen synthase kinase-3, and glucose-6-phosphatase.
- Published
- 2004
16. Discovery of inhibitors of human adipocyte fatty acid-binding protein, a potential type 2 diabetes target
- Author
-
Saba Haile, Stefan Svensson, Lena Rondahl, Tjeerd Barf, Carmen Medina, Fredik Lehmann, Eva Axen, Jonas Uppenberg, and Thomas Lundbaeck
- Subjects
Molecular Sequence Data ,Clinical Biochemistry ,Pharmaceutical Science ,Type 2 diabetes ,Fatty Acid-Binding Proteins ,Biochemistry ,Chemical synthesis ,chemistry.chemical_compound ,Drug Delivery Systems ,Adipocyte ,Drug Discovery ,Adipocytes ,medicine ,Humans ,Amino Acid Sequence ,Molecular Biology ,Binding Sites ,Carbazole ,Binding protein ,Fatty Acids ,Organic Chemistry ,Biological activity ,General Medicine ,medicine.disease ,Fluorescence ,Adipocyte fatty acid binding protein ,Diabetes Mellitus, Type 2 ,chemistry ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,Carrier Proteins ,Fluorescence anisotropy - Abstract
Low micromolar human A-FABP inhibitors were found by utilizing a fluorescence polarization assay, X-ray crystallography and modeling. The carbazole- and indole-based inhibitors displayed approximately 10-fold preferences over human H-FABP and E-FABP, and are highly selective against I-FABP. This communication describes the SAR for drug-like synthetic inhibitors of human A-FABP.
- Published
- 2004
17. Synthesis and Biological Activity of a Novel Class of Small Molecular Weight Peptidomimetic Competitive Inhibitors of Protein Tyrosine Phosphatase 1B
- Author
-
Heinrich J. Schostarez, F. Craig Stevens, John E. Bleasdale, Theresa J. O’Sullivan, Scott D. Larsen, Charlotta Liljebris, Derek Ogg, Barbara J. Palazuk, Tjeerd Barf, and Paul D. May
- Subjects
Models, Molecular ,Peptidomimetic ,Protein tyrosine phosphatase ,Deoxyglucose ,Crystallography, X-Ray ,Receptor tyrosine kinase ,Cell Line ,Structure-Activity Relationship ,Drug Discovery ,Insulin ,Enzyme Inhibitors ,Tyrosine ,Protein Tyrosine Phosphatase, Non-Receptor Type 1 ,biology ,Chemistry ,GRB10 ,Molecular Mimicry ,Hydrogen Bonding ,Dipeptides ,IRS2 ,Molecular Weight ,Insulin receptor ,Biochemistry ,ROR1 ,biology.protein ,Molecular Medicine ,Protein Tyrosine Phosphatases ,Peptides - Abstract
Protein tyrosine phosphatase 1B (PTP1B) negatively regulates insulin signaling in part by dephosphorylating key tyrosine residues within the regulatory domain of the beta-subunit of the insulin receptor (IR), thereby attenuating receptor tyrosine kinase activity. Inhibition of PTP1B is therefore anticipated to improve insulin resistance and has recently become the focus of discovery efforts aimed at identifying new drugs to treat type II diabetes. We previously reported that the tripeptide Ac-Asp-Tyr(SO(3)H)-Nle-NH(2) is a surprisingly effective inhibitor of PTP1B (K(i) = 5 microM). With the goal of improving the stability and potency of this lead, as well as attenuating its peptidic character, an analogue program was undertaken. Specific elements of the initial phase of this program included replacement of the N- and C-termini with non-amino acid components, modification of the tyrosine subunit, and replacement of the tyrosine sulfate with other potential phosphate mimics. The most potent analogue arising from this effort was triacid 71, which inhibits PTP1B competitively with a K(i) = 0.22 microM without inhibiting SHP-2 or LAR at concentrations up to 100 microM. Overall, the inhibitors generated in this work showed little or no enhancement of insulin signaling in cellular assays. However, potential prodrug triester 70 did induce enhancements in 2-deoxyglucose uptake into two different cell lines with concomitant augmentation of the tyrosine phosphorylation levels of insulin-signaling molecules. Key elements of the overall SAR reported herein include confirmation of the effectiveness and remarkable PTP1B-specificity of the novel tyrosine phosphate bioisostere, O-carboxymethyl salicylic acid; demonstration that the tyrosine skeleton is optimal relative to closely related structures; replacement of the p-1 aspartic acid with phenylalanine with little effect on activity; and demonstration that inhibitory activity can be maintained in the absence of an N-terminal carboxylic acid. An X-ray cocrystal structure of an analogue bearing a neutral N-terminus (69) bound to PTP1B is reported that confirms a mode of binding similar to that of peptidic substrates.
- Published
- 2001
18. Investigations with GMC2021 in experimental models predictive of antimigraine activity and coronary side-effect potential
- Author
-
Pramod R. Saxena, Peter De Vries, Jan P.C. Heiligers, Antoinette Maassen VanDenBrink, Willem A. Bax, Tjeerd Barf, and Håkan Wikström
- Subjects
medicine.medical_specialty ,Side effect ,Swine ,medicine.drug_class ,Migraine Disorders ,Hemodynamics ,Internal medicine ,Animals ,Humans ,Vasoconstrictor Agents ,Medicine ,Pharmacology ,Sumatriptan ,business.industry ,Blood flow ,Receptor antagonist ,Coronary Vessels ,Carotid Arteries ,medicine.anatomical_structure ,Anesthesia ,cardiovascular system ,Cardiology ,Ergotamine ,medicine.symptom ,business ,Vasoconstriction ,Artery ,medicine.drug - Abstract
Several acutely acting antimigraine drugs, including sumatriptan and other second generation 5-HT1D receptor agonists, have the ability to constrict porcine carotid arteriovenous anastomoses as well as the human isolated coronary artery. These two experimental models seem to serve as indicators, respectively, for the therapeutic and coronary side-effect potential of the compounds. Using these two models, we have now investigated the effects of GMC2021 (3-[2-(dimethylanimo)ethyl]-5-[(trifluoromethyl)sulfonyl]oxy][1 H]indole oxalate, a close analogue of sumatriptan. GMC2021 (30, 100, 300 and 1000 micrograms.kg-1, i.v.) decreased the total carotid blood flow by exclusively decreasing arteriovenous anastomotic blood flow; capillary blood flow to the skin and ears was moderately increased. The mean +/- S.E.M. dose of GMC2021 eliciting a 50% decrease (ED50) in the porcine carotid arteriovenous anastomotic blood flow was found to be 1.1 +/- 0.3 mumol.kg-1 and the highest dose (1000 micrograms.kg-1) produced a 67 +/- 4% reduction. The carotid haemodynamic effects of GMC2021 were reduced by the selective 5-HT1D receptor antagonist, GR127935 (N-[methoxy-3-(4-methyl-1- piperazinyl)phenyl]-2'-methyl-4'-(5-methyl-1,2,4-oxadiazol-3-yl)[1 , 1-biphenyl]-4-carboxamide hydrochloride), which completely antagonizes porcine carotid haemodynamic responses to sumatriptan (ED50: 0.16 mumol.kg-1, i.v.). Compared to sumatriptan (pD2: 6.12 +/- 0.15; Emax: 31.3 +/- 12.3% of contractions to 100 mM K+), GMC2021 was less potent in constricting the human isolated coronary artery (pD2: 5.45 +/- 0.2; Emax: 21.0 +/- 4.8% of contractions to 100 mM K+). The above results suggest that GMC2021 constricts carotid arteriovenous anastomoses partly by a 5-HT1D receptor and partly by another, probably novel, receptor and that GMC2021 should be able to abort migraine headaches in patients, with perhaps a less propensity for coronary side effects.
- Published
- 1996
19. Characterization of the radioactive metabolites of the 5-HT1A receptor radioligand, [O-methl-11C]WAY-100635, in monkey and human plasma by HPLC: Comparison of the behaviour of an identified radioactive metabolite with parent radioligand in monkey using PET
- Author
-
Julie A. McCarron, Victor W. Pike, Christer Halldin, Carl-Gunnar Swahn, Ian Anthony Cliffe, Paul M. Grasby, Camilla Lundkvist, Sajinder K. Luthra, Tjeerd Barf, Håkan Wikström, Safiye Osman, Allan Fletcher, Christopher J. Bench, Lars Farde, and Natalie Ginovart
- Subjects
Male ,Cancer Research ,Time Factors ,Metabolic Clearance Rate ,Pyridines ,Stereochemistry ,Metabolite ,Piperazines ,chemistry.chemical_compound ,Pharmacokinetics ,In vivo ,medicine ,Radioligand ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Carbon Radioisotopes ,Receptor ,Biotransformation ,5-HT receptor ,Radiochemistry ,Brain ,Human brain ,Kinetics ,Macaca fascicularis ,medicine.anatomical_structure ,chemistry ,Receptors, Serotonin ,Molecular Medicine ,5-HT1A receptor ,Serotonin Antagonists ,Receptors, Serotonin, 5-HT1 ,Tomography, Emission-Computed - Abstract
N-(2-(4-(2-Methoxy-phenyl)-1-piperazin-1-yl)ethyl)-N-(2-pyridyl) cyclohexanecarboxamide (WAY-100635), labelled in the O-methyl group with carbon-11 (t1/2 = 20.4 min), is a promising radioligand for application with positron emission tomography (PET) to the study of 5-HT1A receptors in living human brain. An understanding of the metabolism of this new radioligand is crucial to the development of a biomathematical model for the interpretation of the kinetics of radioactivity uptake in brain in terms of receptor-binding parameters. After intravenous injection of [O-methyl-11C]WAY-100635 into humans, radioactivity was found to clear rapidly from blood and plasma. By using established methods for the analysis of radioactivity in plasma, it was found that intravenously injected [O-methyl-11C]WAY-100635 is rapidly metabolised to more polar radioactive compounds in a cynomolgus monkey and in humans. Thus, at 60 min postinjection, parent radioligand represented 40% and 5% of the radioactivity in monkey and human plasma, respectively. In monkey and human, one of the radioactive metabolites was identified as the descyclohexanecarbonyl analogue of the parent radioligand, namely [O-methyl-11C]WAY-100634. This compound is known to have high affinity for 5-HT1A receptors and alpha 1-adrenoceptors. In a PET experiment it was demonstrated that, after IV injection of [O-methyl-11C]WAY-100634 into a cynomolgus monkey, radioactivity was avidly taken up by brain. Uptake of radioactivity was higher in 5-HT1A receptor-rich frontal cortex than in cerebellum, which is devoid of 5-HT1A receptors. Polar radioactive metabolites appeared in plasma. The results suggest that the use of WAY-100635 labelled with carbon-11 in its cyclohexanecarbonyl moiety may provide enhanced signal contrast in PET studies and a possibility to develop a simple biomathematical model for regional brain radioactivity uptake.
- Published
- 1996
20. Novel ATP competitive MK2 inhibitors with potent biochemical and cell-based activity throughout the series
- Author
-
Arthur Oubrie, Carsten Schultz-Fademrecht, Vera de Kimpe, Bas van de Kar, Tjeerd Barf, Bert Kazemier, Niels Hoogenboom, Rianne Goorden, Judith Borsboom, Edwin de Zwart, Allard Kaptein, Michiel Scheffers, Jos Lommerse, Jeroen A.D.M. de Roos, Ruud van der Heijden, and Maaike van Hoek
- Subjects
medicine.medical_treatment ,Chemistry, Pharmaceutical ,Clinical Biochemistry ,HSP27 Heat-Shock Proteins ,Pharmaceutical Science ,Protein Serine-Threonine Kinases ,Crystallography, X-Ray ,Biochemistry ,chemistry.chemical_compound ,Hsp27 ,Heat shock protein ,Drug Discovery ,medicine ,Potency ,Humans ,Computer Simulation ,Pyrroles ,Phosphorylation ,Benzamide ,Molecular Biology ,Protein Kinase Inhibitors ,Heat-Shock Proteins ,Piperidones ,biology ,Tumor Necrosis Factor-alpha ,MAPKAPK2 ,Organic Chemistry ,Intracellular Signaling Peptides and Proteins ,Cytokine ,chemistry ,Models, Chemical ,Drug Design ,Benzamides ,biology.protein ,Molecular Medicine ,Cytokines ,Tumor necrosis factor alpha ,Molecular Chaperones - Abstract
Optimization of our previously described pyrrolopiperidone series led to the identification of a new benzamide sub-series, which exhibits consistently high potency in biochemical and cell-based assays throughout the series. Strong inhibition of LPS-induced production of the cytokine TNFα is coupled to the regulation of HSP27 phosphorylation, indicating that the observed cellular effects result from the inhibition of MK2. X-ray crystallographic and computational analyses provide a rationale for the high potency of the series.
- Published
- 2011
21. Discovery of selective and orally available spiro-3-piperidyl ATP-competitive MK2 inhibitors
- Author
-
Judith Versteegh, Joeri Johannes Petrus De Wit, Arthur Oubrie, Niels Hoogenboom, Tjeerd Barf, Maaike van Hoek, Bert Kazemier, Allard Kaptein, Carsten Schultz-Fademrecht, Paula Leandro-Garcia, Frank Wijnands, John Dulos, Martin Jaeger, Bas van de Kar, Jeroen A.D.M. de Roos, Gerard Mt Vogel, Mario van Zeeland, Edwin de Zwart, Vera de Kimpe, and Judith Borsboom
- Subjects
Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Administration, Oral ,Biological Availability ,Stereoisomerism ,Protein Serine-Threonine Kinases ,Crystallography, X-Ray ,Biochemistry ,Chemical synthesis ,Binding, Competitive ,Substrate Specificity ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,Piperidines ,Drug Discovery ,Structure–activity relationship ,Moiety ,Animals ,Humans ,Spiro Compounds ,Molecular Biology ,Protein Kinase Inhibitors ,Chromatography, High Pressure Liquid ,chemistry.chemical_classification ,biology ,Molecular Structure ,Drug discovery ,Organic Chemistry ,Intracellular Signaling Peptides and Proteins ,Bioavailability ,Rats ,Enzyme Activation ,Disease Models, Animal ,Enzyme ,chemistry ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,Caco-2 Cells - Abstract
The identification of a potent, selective, and orally available MK2 inhibitor series is described. The initial absence of oral bioavailability was successfully tackled by moving the basic nitrogen of the spiro-4-piperidyl moiety towards the electron-deficient pyrrolepyridinedione core, thereby reducing the pK(a) and improving Caco-2 permeability. The resulting racemic spiro-3-piperidyl analogues were separated by chiral preparative HPLC, and the activity towards MK2 inhibition was shown to reside mostly in the first eluting stereoisomer. This led to the identification of new MK2 inhibitors, such as (S)-23, with low nanomolar biochemical inhibition (EC(50) 7.4 nM) and submicromolar cellular target engagement activity (EC(50) 0.5 μM).
- Published
- 2011
22. Structure-based lead identification of ATP-competitive MK2 inhibitors
- Author
-
Judith Versteegh, Nicole Seegers, Henri Klop, Tjeerd Barf, Claudia Hofstra, Jeroen A.D.M. de Roos, Carsten Schultz-Fademrecht, Mario van Zeeland, Arthur Oubrie, Bert Kazemier, Maaike van Hoek, Dennis Demont, Allard Kaptein, Jorrit J. Hornberg, Sander de Wilde, Ruben L. Smeets, Bas van de Kar, Ruud van der Heijden, and Richard van Someren
- Subjects
Drug ,Models, Molecular ,media_common.quotation_subject ,Clinical Biochemistry ,Pharmaceutical Science ,Computational biology ,Protein Serine-Threonine Kinases ,Crystallography, X-Ray ,Auto-immunity, transplantation and immunotherapy [N4i 4] ,Biochemistry ,Binding, Competitive ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,Adenosine Triphosphate ,In vivo ,Drug Discovery ,Animals ,Humans ,Molecular Biology ,Protein Kinase Inhibitors ,ADME ,media_common ,chemistry.chemical_classification ,biology ,Molecular Structure ,Drug discovery ,Kinase ,MAPKAPK2 ,Organic Chemistry ,Intracellular Signaling Peptides and Proteins ,Rats ,Disease Models, Animal ,Enzyme ,chemistry ,Enzyme inhibitor ,Drug Design ,biology.protein ,Molecular Medicine ,Caco-2 Cells - Abstract
Item does not contain fulltext MK2 kinase is a promising drug discovery target for the treatment of inflammatory diseases. Here, we describe the discovery of novel MK2 inhibitors using X-ray crystallography and structure-based drug design. The lead has in vivo efficacy in a short-term preclinical model.
- Published
- 2011
23. N-Benzyl-indolo carboxylic acids: Design and synthesis of potent and selective adipocyte fatty-acid binding protein (A-FABP) inhibitors
- Author
-
Kristin Hammer, Jonas Uppenberg, Stefan Svensson, Eva Axen, Saba Haile, Carmen Medina, Thomas Lundbäck, Tjeerd Barf, Lena Rondahl, and Fredrik Lehmann
- Subjects
medicine.drug_class ,Stereochemistry ,Carboxylic acid ,Chemistry, Pharmaceutical ,Clinical Biochemistry ,Carboxylic Acids ,Pharmaceutical Science ,Carboxamide ,Crystallography, X-Ray ,Fatty Acid-Binding Proteins ,Biochemistry ,Chemical synthesis ,Fatty acid-binding protein ,chemistry.chemical_compound ,Inhibitory Concentration 50 ,Mice ,Adipocyte ,Drug Discovery ,medicine ,Adipocytes ,Animals ,Humans ,Molecular Biology ,chemistry.chemical_classification ,Indole test ,Binding Sites ,Molecular Structure ,Binding protein ,Macrophages ,Organic Chemistry ,Small molecule ,Spectrometry, Fluorescence ,chemistry ,Models, Chemical ,Drug Design ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) - Abstract
Small molecule inhibitors of adipocyte fatty-acid binding protein (A-FABP) have gained renewed interest following the recent publication of pharmacologically beneficial effects of such inhibitors. Despite the potential utility of selective A-FABP inhibitors within the fields of metabolic disease, inflammation and atherosclerosis, there are few examples of useful A-FABP inhibitors in the public domain. Herein, we describe the optimization of N-benzyl-tetrahydrocarbazole derivatives through the use of co-crystal structure guided medicinal chemistry efforts. This led to the identification of a potent and selective class of A-FABP inhibitors as illustrated by N-benzyl-hexahydrocyclohepta[b]indole 30.
- Published
- 2008
24. Abstract 2596: ACP-196: a novel covalent Bruton's tyrosine kinase (Btk) inhibitor with improved selectivity and in vivo target coverage in chronic lymphocytic leukemia (CLL) patients
- Author
-
Elena Bibikova, Ahmed Hamdy, Edwin de Zwart, Fanny Krantz, Raquel Izumi, Allard Kaptein, Michael Gulrajani, Tjeerd Barf, Bart Van Lith, Todd Covey, and Bas van de Kar
- Subjects
CD20 ,Cancer Research ,biology ,business.industry ,Kinase ,Chronic lymphocytic leukemia ,Pharmacology ,medicine.disease ,chemistry.chemical_compound ,Oncology ,chemistry ,In vivo ,Ibrutinib ,biology.protein ,medicine ,Bruton's tyrosine kinase ,Mantle cell lymphoma ,business ,Tyrosine kinase - Abstract
Ibrutinib, a first generation Btk inhibitor, is approved for the treatment of CLL and mantle cell lymphoma; known toxicities include atrial fibrillation, diarrhea, rash, arthralgia and bleeding events (1). Recent reports show ibrutinib's off target effects may negatively impact its potential for combined therapy with anti-CD20 antibodies (2,3). Here we describe the pharmacologic characterization of ACP-196 a potent, novel second generation Btk inhibitor, which binds covalently to Cys481 with improved selectivity and in vivo target coverage. Compared to ibrutinib and CC-292, ACP-196 demonstrated higher selectivity for Btk when profiled against a panel of 395 non-mutant kinases (1 μM) in a competitive binding assay. IC50 determinations on 9 kinases with a Cys in the same position as Btk showed ACP-196 to be the most selective. The improved selectivity is related to the reduced intrinsic reactivity of ACP-196's electrophile. Importantly, unlike ibrutinib, ACP-196 did not inhibit EGFR, Itk or Txk. Phosphoflow assays on EGFR expressing cell lines confirmed ibrutinib's EGFR inhibition (EC50: 47-66 nM) with no inhibition observed for ACP-196 at 10 μM. These data may explain the ibrutinib-related incidence of diarrhea and rash. Ibrutinib's potency on Itk and Txk may explain why it interferes with cell-mediated anti-tumor activities of therapeutic CD20 antibodies and immune-mediated killing in the tumor microenvironment (2,3). In human whole blood, ACP-196 and ibrutinib showed robust and equipotent inhibitory activity on B-cell receptor induced responses in the low nM range, whereas CC-292 was 10-20 fold less potent. In vivo, oral administration of ACP-196 in mice resulted in dose-dependent inhibition of anti-IgM-induced CD86 expression in CD19+ splenocytes with an ED50 of 0.34 mg/kg compared to 0.91 mg/kg for ibrutinib. A similar model was used to compare the duration of Btk inhibition after a single oral dose of 25 mg/kg. ACP-196 and ibrutinib inhibited CD86 expression >90% at 3h and ∼50% at 24h postdose. In contrast, CC-292 inhibited ∼50% at 3h and ∼20% at 24h postdose. An ELISA based Btk target occupancy assay was developed to measure target coverage in preclinical and clinical studies. In healthy volunteers, ACP-196 at an oral dose of 100 mg QD showed >90% target coverage over a 24h period. Btk occupancy and regulation of the PD markers (CD69 and CD86) correlated with PK parameters for exposure. In CLL patients, after 7 days of dosing with ACP-196 at 200 mg QD, 94% Btk target occupancy was observed compared with ∼80% reported for ibrutinib at 420 mg QD (4). In conclusion, ACP-196 is a novel Btk inhibitor with key pharmacologic differentiators versus ibrutinib and CC-292. ACP-196 is currently being evaluated in clinical trials. 1. IMBRUVICA package insert 2014 2. Rajasekaran Blood 2014 Abstr # 3118 3. Da Roit Haematologica 2014 4. Byrd NEJM 2013 Citation Format: Todd Covey, Tjeerd Barf, Michael Gulrajani, Fanny Krantz, Bart van Lith, Elena Bibikova, Bas van de Kar, Edwin de Zwart, Ahmed Hamdy, Raquel Izumi, Allard Kaptein. ACP-196: a novel covalent Bruton's tyrosine kinase (Btk) inhibitor with improved selectivity and in vivo target coverage in chronic lymphocytic leukemia (CLL) patients. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2596. doi:10.1158/1538-7445.AM2015-2596
- Published
- 2015
25. Active site variability of type 1 11beta-hydroxysteroid dehydrogenase revealed by selective inhibitors and cross-species comparisons
- Author
-
Stefan Svensson, Margareta Forsgren, Doreen Mitschke, Naeem Shafqat, Malin Hult, Tjeerd Barf, Udo Oppermann, Lars Abrahmsén, Jerk Vallgårda, and Björn Elleby
- Subjects
Protein Conformation ,Guinea Pigs ,Molecular Sequence Data ,Dehydrogenase ,Biology ,Biochemistry ,Binding, Competitive ,Mice ,Structure-Activity Relationship ,Endocrinology ,Dogs ,Cricetinae ,11-beta-Hydroxysteroid Dehydrogenase Type 1 ,Animals ,Humans ,Amino Acid Sequence ,Binding site ,Enzyme Inhibitors ,Molecular Biology ,chemistry.chemical_classification ,Sulfonamides ,Binding Sites ,Protein primary structure ,Active site ,Hydroxysteroid Dehydrogenases ,Ligand (biochemistry) ,Rats ,Transmembrane domain ,Thiazoles ,Enzyme ,chemistry ,biology.protein ,Cats - Abstract
The NADPH-dependent enzyme type 1 11beta-hydroxysteroid dehydrogenase (11beta-HSD1) activates in a tissue-specific manner circulating pro-glucocorticoid hormones (cortisone in humans) to the 11beta-OH ligand (cortisol in humans), which is able to bind to its cognate receptor and regulate gene transcription. Modulation of this pre-receptor activation mechanism by selective enzyme inhibitors is a desirable goal in the treatment of insulin resistance and related metabolic disorders. Like most other hydroxysteroid dehydrogenases 11beta-HSD1 belongs to the evolutionarily conserved enzyme superfamily of short-chain dehydrogenases/reductases (SDR). The enzyme is anchored within the endoplasmic reticulum through an N-terminal transmembrane domain. In this study we aimed to characterize the active site of mammalian 11beta-HSD1 by determining primary structures from several mammalian lines (cat, hamster, cynomolgus, chimpanzee, dog) thus increasing substantially available sequence information, and allowing us to determine highly variable and constant parts within the primary structure. These regions were mapped to the recently determined three-dimensional structure and are mostly found around the substrate binding site. Furthermore we performed inhibition studies by using different series of inhibitors, comprising 11beta-HSD1 selective arylsulfonamidothiazoles and the unselective steroid-based compound carbenoxolone. The different arylsulfonamidothiazoles display distinct inhibition profiles versus the mammalian species tested, with several tight binding inhibitors for the human enzyme (Ki approximately 50 nM), intermediate for mouse, and weak or not binding inhibitors for rat and guinea pig (Ki>3 microM). Analysis of the inhibition mode reveals that the tight binding inhibitor BVT.528 is a competitive inhibitor for the human form, whereas the related compound BVT.2733 displays a mixed-type inhibition pattern versus the mouse enzyme. Taken together, this structure-activity study provides increased insight into active site complexity and catalytic mechanism of 11beta-HSD1, useful for further inhibitor design.
- Published
- 2006
26. Substituted Benzylamino-6-(trifluoromethyl)pyrimidin-4(1H)-ones: A Novel Class of Selective Human A-FABP Inhibitors
- Author
-
Jonas Uppenberg, Tjeerd Barf, Eva Axen, Lena Rondahl, Thomas Lundbäck, and Rune Ringom
- Subjects
Benzylamines ,Class (set theory) ,Pyrimidine ,Pyridines ,Stereochemistry ,Clinical Biochemistry ,Substituent ,Pharmaceutical Science ,Ring (chemistry) ,Fatty Acid-Binding Proteins ,Biochemistry ,Chemical synthesis ,chemistry.chemical_compound ,Drug Discovery ,Adipocytes ,Humans ,Molecular Biology ,Biological evaluation ,Trifluoromethyl ,Chemistry ,Organic Chemistry ,General Medicine ,Chain length ,Pyrimidines ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,Selectivity ,Carrier Proteins - Abstract
The synthesis and biological evaluation of novel human A-FABP inhibitors based on the 6-(trifluoromethyl)pyrimidine-4(1H)-one scaffold is described. Two series of compounds, bearing either an amino or carbon substituent in the 2-position of the pyrimidine ring were investigated. Modification of substituents and chain length optimization led to novel compounds with low micromolar activity and good selectivity for human A-FABP.
- Published
- 2004
27. Arylsulfonamidothiazoles as a new class of potential antidiabetic drugs. Discovery of potent and selective inhibitors of the 11beta-hydroxysteroid dehydrogenase type 1
- Author
-
Rolf Olsson, Charlotta Häggström, Lars O. M. Engblom, Guido Kurz, Vivienne Larwood, Kent Axelsson, Naimie H. M. Edling, Birgitta Ohman, Tjeerd Barf, Yuko Rönquist-Nii, Lars Abrahmsén, Rikard Emond, Erifili Mosialou, Alf Nygren, Jerk Vallgarda, and Peteris Alberts
- Subjects
Blood Glucose ,medicine.drug_class ,Dehydrogenase ,Carboxamide ,Isozyme ,Mice ,Structure-Activity Relationship ,11β-hydroxysteroid dehydrogenase type 1 ,Drug Discovery ,11-beta-Hydroxysteroid Dehydrogenase Type 1 ,medicine ,Structure–activity relationship ,Animals ,Humans ,Hypoglycemic Agents ,chemistry.chemical_classification ,Sulfonamides ,biology ,Hydroxysteroid Dehydrogenases ,Biological activity ,Thiazoles ,Enzyme ,chemistry ,Biochemistry ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,hormones, hormone substitutes, and hormone antagonists - Abstract
Novel antidiabetic arylsulfonamidothiazoles are presented that exert action through selective inhibition of the 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) enzyme, thereby attenuating hepatic gluconeogenesis. The diethylamide derivative 2a was shown to potently inhibit human 11beta-HSD1 (IC(50) = 52 nM), whereas the N-methylpiperazinamide analogue 2b only inhibited murine 11beta-HSD1 (IC(50) = 96 nM). Both compounds showed >200-fold selectivity over human and murine 11beta-HSD2. 2b was subsequently shown to reduce glucose levels in diabetic KKA(y) mice, substantiating the 11beta-HSD1 enzyme as a target for the treatment of type 2 diabetes.
- Published
- 2002
28. Abstract 2624: The novel Bruton's tyrosine kinase inhibitor ACP-196 shows in vivo efficacy against human chronic lymphocytic leukemia cells xenografted to the NSG mouse model
- Author
-
Dolors Colomer, Tjeerd Barf, Arnau Montraveta, Adrian Wiestner, Carsten Utoft Niemann, Tim Ingallinera, and Sarah E. M. Herman
- Subjects
Cancer Research ,biology ,medicine.drug_class ,business.industry ,Chronic lymphocytic leukemia ,B-cell receptor ,medicine.disease ,Tyrosine-kinase inhibitor ,Leukemia ,chemistry.chemical_compound ,Oncology ,chemistry ,hemic and lymphatic diseases ,Ibrutinib ,Immunology ,Cancer research ,NSG mouse ,biology.protein ,medicine ,Bruton's tyrosine kinase ,business ,Tyrosine kinase - Abstract
Background: Targeting Bruton's tyrosine kinase (BTK), an essential kinase in the B cell receptor (BCR) pathway in patients with chronic lymphocytic leukemia (CLL) has proven very effective. For the first generation BTK inhibitor ibrutinib, clinical response rates >70% and 75% progression free survival >2 years have been reported for previously treated patients (Byrd, NEJM, 2013). ACP-196 is a novel, second generation, irreversible BTK inhibitor that may show advantages in terms of binding specificity and drug-drug interactions compared to the first generation BTK inhibitor. Survival and proliferation of CLL cells is highly dependent on microenvironment interaction, which must be taken into consideration when testing new drugs for CLL. We have previously shown that human CLL cells engrafted in the spleen of NSG mice have comparable tumor biology to human lymph node resident CLL cells (Herman, Leukemia, 2013). We here demonstrate the in vivo effects of ACP-196 monotherapy against CLL cells in the NSG xenograft model. Methods: Peripheral blood mononuclear cells from previously untreated CLL patients were injected intravenously into NSG mice. Mice received ACP-196 through the drinking water. The effect of ACP-196 on xenografted CLL cells from peripheral blood and spleen was assessed by flow cytometry. Results: At all dose levels tested, ACP-196 significantly inhibited proliferation of human CLL cells in the spleens of NSG mice, as measured by Ki67 expression (P=0.002). The mean Ki67 decrease was 58%, 70% and 73%, respectively for the low, intermediate, and high dose level compared with vehicle. Tumor burden in spleens from mice treated with the high dose of ACP-196 decreased compared with the vehicle treated group (P=0.007). As seen with other BCR inhibitors, ACP-196 transiently increased CLL cell counts in the peripheral blood in a dose dependent manner (p=0.01). ACP-196 inhibited BCR signaling in vivo, as demonstrated by reduced phosphorylation of PLCγ2. Conclusions: Results presented here for ACP-196 are in accordance with the expected profile of an irreversible BTK inhibitor. The NSG CLL xenograft model will be used for comparative testing of different BTK inhibitors and exploration of combination therapies. Citation Format: Carsten U. Niemann, Arnau Montraveta, Sarah E. M. Herman, Tim Ingallinera, Tjeerd Barf, Dolors Colomer, Adrian Wiestner. The novel Bruton's tyrosine kinase inhibitor ACP-196 shows in vivo efficacy against human chronic lymphocytic leukemia cells xenografted to the NSG mouse model. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2624. doi:10.1158/1538-7445.AM2014-2624
- Published
- 2014
29. 5-(sulfonyl)oxy-tryptamines and ethylamino side chain restricted derivatives. Structure-affinity relationships for h5-HT1B and h5-HT1D receptors
- Author
-
S. Sundell, Christiane Palmier, Petrus J. Pauwels, Tjeerd Barf, Stéphanie Tardif, Max Lundmark, Håkan Wikström, Groningen Research Institute of Pharmacy, and Faculty of Science and Engineering
- Subjects
Tryptamine ,AUTORECEPTORS ,Magnetic Resonance Spectroscopy ,Stereochemistry ,POTENT AGONISTS ,Clinical Biochemistry ,5-HT ,Pharmaceutical Science ,Crystallography, X-Ray ,Biochemistry ,Mass Spectrometry ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,BINDING ,Side chain ,sulfonate ,Animals ,Humans ,Molecular Biology ,PHARMACOLOGY ,5-HT receptor ,Sulfonyl ,chemistry.chemical_classification ,SITES ,Trifluoromethyl ,Chemistry ,Organic Chemistry ,Affinities ,Recombinant Proteins ,Tryptamines ,serotonin ,BRAIN MEMBRANES ,h5-HT1D ,h5-HT1B ,5-HT1D RECEPTOR ,SEROTONERGIC ACTIVITY ,Receptor, Serotonin, 5-HT1D ,Receptors, Serotonin ,COS Cells ,SUBFAMILY ,Receptor, Serotonin, 5-HT1B ,Molecular Medicine ,Amine gas treating ,SUMATRIPTAN ,HeLa Cells ,Protein Binding - Abstract
A number of sulfonic acid ester derivatives of serotonin (5-hydroxytryptamine; 5-HT; 1) were prepared and their affinities are compared to that of the reference compound 5-[[(trifluoromethyl)sulfonyl]oxy]-tryptamine (8b). The structure-affinity relationship (SAFIR) is discussed in terms of in vitro binding for cloned human h5-HT1A, h5-HT1B and h5-HT1D receptors. All tryptamine derivatives exhibited the best affinities for h5-HT1D receptors but still, these were comparatively lower than that of compound 8b. 5-Tosylated tryptamine 11b (K-i = 6 nM) and the sulfamate derivatives 13b and 14b (K-i = 7 and 11 nM, respectively) were found to have the highest affinities for the h5-HT1D receptor. Other tryptamine derivatives displayed moderate binding for h5-HT1A and h5-HT1B receptors, along with Ki values ranging from 14-20 nM for the h5-HT1D sites. In addition, the syntheses of two alkylamino side chain restricted derivatives are described. 3-Amino-6-[[(trifluoromethyl)sulfonyl]oxy]-1,2,3,4-tetrahydrocarbazole 21, as well as 4-[5-[[(trifluoromethyl)sulfonyl]oxy]-1H-indol-3-yl]piperidines 24 and 25, induced a shift in selectivity in favor of the h5-HT1B receptor. The relatively longer distance between the basic amine and a hydrogen-bond accepting oxygen in 21, 24 and 25 as compared to the non-restricted tryptamines, is likely responsible for this observation. (C) 1998 Elsevier Science Ltd. All rights reserved.
- Published
- 1998
30. Potential anxiolytic properties of R-(+)-8-OSO2CF3-PAT, a 5-HT 1A receptor agonist
- Author
-
Béla Bohus, Geert Damsma, Clas Sonesson, Håkan Wikström, S. Mechiel Korte, Gerdien A.H. Korte-Bouws, and Tjeerd Barf
- Subjects
Agonist ,5-HIAA/5-HT (5-hydroxy-indole acetic acid 5-hydroxytryptamine) ratio ,Male ,medicine.medical_specialty ,defensive burying ,AUTORECEPTORS ,Serotonin ,medicine.drug_class ,inescapable footshock ,RAT-BRAIN ,Anxiolytic ,Buspirone ,chemistry.chemical_compound ,BUSPIRONE ,Internal medicine ,medicine ,ANXIETY ,DRUGS ,Animals ,5-HT1A receptor ,Rats, Wistar ,5-HT receptor ,BURYING TEST ,Pharmacology ,DORSAL ,8-Hydroxy-2-(di-n-propylamino)tetralin ,RAPHE NUCLEI ,Behavior, Animal ,Chemistry ,8-OH-DPAT ,Brain ,Hydroxyindoleacetic Acid ,plus-maze ,Rats ,Serotonin Receptor Agonists ,Endocrinology ,Anti-Anxiety Agents ,Autoreceptor ,medicine.drug - Abstract
The anxiolytic property of R-(+)-8-OSO3CF3-PAT (R-(+)-8-[[(trifluoromethyl)sulfonyl]oxy]-2-(n-propyl-amino)tetralin), a 5-HT1A receptor agonist, was evaluated in Wistar rats by means of animal models of anxiety, the conditioned defensive burying model and the conditioned stress-induced freezing response followed by the elevated plus-maze tear, respectively, In addition, the 5-HIAA/5-HT ratio (5-hydroxy-indole acetic acid/5-hydroxytryptamine) of rat brain homogenates was studied. Acute drug administration resulted in abolition of the burying behaviour (3 mg/kg i.p.), a dose-dependent decrease of rearing and induction of hyperphagia. R-(+)8-OSO2CF3-PAT had no effect on conditioned footshock-induced freezing behaviour but increased open-arm activity in the rats on the plus-maze, The 5-HIAA/5-HT ratio was decreased in the lateral septum (1 and 3 mg/kg), dorsal hippocampus (3 mg/kg) and somatosensory cortex (3 mg/kg), implying that R-(+)-8-OSO2CF3-PAT affects particularly the limbic system in anxiety-inducing situations.
- Published
- 1996
31. Self-assembling chiral metallo-clefts: Synthesis and molecular structure of N,N'-bis(12H-benzo[a]xanthen-12-ylidene)-1,2-ethanediamine zinc(II)-dichloride complex
- Author
-
Ben L. Feringa, Tjeerd Barf, Anthony L. Spek, Fré van Bolhuis, Johan F.G.A. Jansen, and Synthetic Organic Chemistry
- Subjects
Ligand ,Stereochemistry ,Hydrosilylation ,Imine ,Thio ,chemistry.chemical_element ,General Chemistry ,Zinc ,Medicinal chemistry ,Chloride ,chemistry.chemical_compound ,chemistry ,medicine ,Molecule ,Isomerization ,medicine.drug - Abstract
Bulky ligands for cleft-type metal complexes were synthesized from thio ketones and diamines in yields varying from 20-80%. A molecular structure determination of one ligand N,N'-bis(12H-benzo[a]-xanthen-12-ylidene)-1,2-propanediamine (19) was performed. A metallo-cleft was constructed by a self-assembling process with zinc(II) chloride. A crystallographic study of this zinc complex (24) revealed surprisingly that an anti-syn inversion of the imine moieties of the ligand had taken place during complexation.
- Published
- 1993
32. Inside Cover: Characterization of Irreversible Kinase Inhibitors by Directly Detecting Covalent Bond Formation: A Tool for Dissecting Kinase Drug Resistance (ChemBioChem 18/2010)
- Author
-
Daniel Rauh, Matthias Rabiller, Hans C.A. Raaijmakers, Sabine Klüter, Willem A. L. van Otterlo, Christian Grütter, Vijaykumar Pawar, Jeffrey R. Simard, Tjeerd Barf, and Haridas B. Rode
- Subjects
Covalent bond ,Kinase ,Chemistry ,Organic Chemistry ,Molecular Medicine ,Cover (algebra) ,Drug resistance ,Molecular Biology ,Biochemistry ,Combinatorial chemistry ,Fluorescence spectroscopy ,Characterization (materials science) - Published
- 2010
33. Recent progress in 11-beta-hydroxysteroid dehydrogenase type 1 (11-beta-HSD1) inhibitor development
- Author
-
Meredith Williams and Tjeerd Barf
- Subjects
chemistry.chemical_classification ,medicine.medical_specialty ,Stimulation ,Dehydrogenase ,Disease ,Biology ,medicine.disease ,Endocrinology ,Enzyme ,Glucocorticoid receptor ,chemistry ,Diabetes mellitus ,Internal medicine ,medicine ,Metabolic syndrome ,hormones, hormone substitutes, and hormone antagonists ,Dyslipidemia - Abstract
Glucocorticoids regulate a plethora of processes via stimulation of nuclear glucocorticoid receptors. Exaggerated effects of glucocorticoids can give rise to multiple clinical features typified by Cushing's disease, a metabolic syndrome comprising hyperglycemia, insulin-resistant diabetes, obesity, dyslipidemia and hypertension. Since the enzyme 11-β-hydroxysteroid dehydrogenase type 1 (11-β-HSD1) locally regenerates active glucocorticoids, tissue-specific inhibitors of 11-β-HSD1 have potential for ameliorating the above conditions. This review describes the racent efforts to identify potent and selective 11-β-HSD1 inhibitors, aimed at correcting abnormalities associated with the metabolic syndrome. The primary focus is on aspects of the metabolic syndrome, but a is recognized that inactivation of 11-β-HSD1 may also be beneficial in other diseases.
- Published
- 2006
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.