1
|
Kaul A, Hope H, Xu C, Basavalingappa R, Binz SK, Boily C, Bradley Z, Burt D, Emanuel C, Fairchild J, Egan S, Hildebrand A, Howell V, Huang H, Huff E, Iken A, Knapik S, Lawrence M, Lin H, Lu JW, Mattingly J, McGraw D, McGraw N, Mnich S, Morton W, Ortmann R, Piccinni-Ash T, Saer R, da Silva CS, Stillwell L, Taylor W, Warner E, Wrightstone A, Jacobsen EJ, Anderson DR, Monahan J. Characterization of the dual ITK/JAK3 small molecule covalent inhibitor ATI-2138. J Pharmacol Exp Ther 2025; 392:100054. [PMID: 40023606 DOI: 10.1016/j.jpet.2024.100054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/12/2024] [Indexed: 03/04/2025] Open
Abstract
Aclaris Therapeutics Inc (ATI)-2138 is a novel investigational covalent inhibitor of interleukin-2-inducible T cell kinase (ITK), resting lymphocyte kinase, and Janus kinase 3 (JAK3) in development for the treatment of autoimmune and inflammatory diseases. In this study, we evaluated the inhibitory effects of ATI-2138 on ITK and JAK3 signaling in cells and preclinical animal models and assessed the safety, tolerability, pharmacokinetics, and pharmacodynamics of ATI-2138 in healthy human participants. ATI-2138 potently, selectively, and irreversibly inhibited ITK, resting lymphocyte kinase, and JAK3 kinases with similar potency observed against ITK and JAK3 in biochemical and immune cell signaling assays. Translation from cellular and whole blood studies to in vivo models was observed, wherein ATI-2138 demonstrated disease-modifying activity in 2 rodent models of arthritis and an adoptive T cell model of colitis. In single and multiple ascending dose studies in healthy human participants, ATI-2138 had a favorable safety profile with linear pharmacokinetics. Biomarkers linked to both ITK and JAK3 activity were inhibited with ATI-2138 in an exposure-, dose-, and time-dependent manner and correlated with enzyme, cellular, whole blood, and rodent studies, thereby demonstrating predictive translational properties. As a potential first-in-class dual inhibitor of ITK and JAK3, ATI-2138 may be useful in the treatment of immunoinflammatory diseases. SIGNIFICANCE STATEMENT: Aclaris Therapeutics Inc (ATI)-2138 is a novel covalent inhibitor of interleukin-2-inducible T cell kinase (ITK)/resting lymphocyte kinase and Janus kinase 3 (JAK3). ATI-2138 inhibits JAK3 and ITK in enzyme and functional cellular assays, demonstrates disease-modifying activity in rodent models of arthritis and colitis, and inhibits biomarkers linked to both ITK and JAK3 activity in healthy human participants. With this dual kinase activity against components of these inflammatory signaling pathway, ATI-2138 has the potential for enhanced therapeutic efficacy in the treatment of autoimmune and chronic inflammatory disease.
Collapse
Affiliation(s)
- Aparna Kaul
- Confluence Discovery Technologies, St. Louis, Missouri
| | - Heidi Hope
- Confluence Discovery Technologies, St. Louis, Missouri
| | - Canxin Xu
- Confluence Discovery Technologies, St. Louis, Missouri
| | | | - Sara K Binz
- Confluence Discovery Technologies, St. Louis, Missouri
| | - Chad Boily
- Aclaris Therapeutics, Inc, Wayne, Pennsylvania
| | | | - David Burt
- Aclaris Therapeutics, Inc, Wayne, Pennsylvania
| | | | | | - Sarah Egan
- Confluence Discovery Technologies, St. Louis, Missouri
| | | | | | - Huiyan Huang
- Confluence Discovery Technologies, St. Louis, Missouri
| | - Emma Huff
- Confluence Discovery Technologies, St. Louis, Missouri
| | - Abbygail Iken
- Confluence Discovery Technologies, St. Louis, Missouri
| | | | | | - Huawen Lin
- Confluence Discovery Technologies, St. Louis, Missouri
| | | | | | - Dean McGraw
- Confluence Discovery Technologies, St. Louis, Missouri
| | - Nancy McGraw
- Confluence Discovery Technologies, St. Louis, Missouri
| | - Stephen Mnich
- Confluence Discovery Technologies, St. Louis, Missouri
| | | | | | | | - Rafael Saer
- Confluence Discovery Technologies, St. Louis, Missouri
| | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Kojima K, Watanabe K, Kawai M, Yagi S, Kaku K, Ikenouchi M, Sato T, Kamikozuru K, Yokoyama Y, Takagawa T, Shimizu M, Shinzaki S. Real-world efficacy and safety of tofacitinib treatment in Asian patients with ulcerative colitis. World J Gastroenterol 2024; 30:1871-1886. [PMID: 38659488 PMCID: PMC11036499 DOI: 10.3748/wjg.v30.i13.1871] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 01/30/2024] [Accepted: 03/13/2024] [Indexed: 04/03/2024] Open
Abstract
BACKGROUND Real-world data on tofacitinib (TOF) covering a period of more than 1 year for a sufficient number of Asian patients with ulcerative colitis (UC) are scarce. AIM To investigate the long-term efficacy and safety of TOF treatment for UC, including clinical issues. METHODS We performed a retrospective single-center observational analysis of 111 UC patients administered TOF at Hyogo Medical University as a tertiary inflammatory bowel disease center. All consecutive UC patients who received TOF between May 2018 and February 2020 were enrolled. Patients were followed up until August 2020. The primary outcome was the clinical response rate at week 8. Secondary outcomes included clinical remission at week 8, cumulative persistence rate of TOF administration, colectomy-free survival, relapse after tapering of TOF and predictors of clinical response at week 8 and week 48. RESULTS The clinical response and remission rates were 66.3% and 50.5% at week 8, and 47.1% and 43.5% at week 48, respectively. The overall cumulative clinical remission rate was 61.7% at week 48 and history of anti-tumor necrosis factor-alpha (TNF-α) agents use had no influence (P = 0.25). The cumulative TOF persistence rate at week 48 was significantly lower in patients without clinical remission than in those with remission at week 8 (30.9% vs 88.1%; P < 0.001). Baseline partial Mayo Score was significantly lower in responders vs non-responders at week 8 (odds ratio: 0.61, 95% confidence interval: 0.45-0.82, P = 0.001). Relapse occurred in 45.7% of patients after TOF tapering, and 85.7% of patients responded within 4 wk after re-increase. All 6 patients with herpes zoster (HZ) developed the infection after achieving remission by TOF. CONCLUSION TOF was more effective in UC patients with mild activity at baseline and its efficacy was not affected by previous treatment with anti-TNF-α agents. Most relapsed patients responded again after re-increase of TOF and nearly half relapsed after tapering off TOF. Special attention is needed for tapering and HZ.
Collapse
Affiliation(s)
- Kentaro Kojima
- Department of Gastroenterology, Hyogo Medical University, Nishinomiya 663-8501, Japan
- Department of Gastroenterology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Kenji Watanabe
- Department of Gastroenterology, Hyogo Medical University, Nishinomiya 663-8501, Japan
- Department of Internal Medicine for Inflammatory Bowel Disease, The University of Toyama, Toyama 930-0194, Japan
| | - Mikio Kawai
- Department of Gastroenterology, Hyogo Medical University, Nishinomiya 663-8501, Japan
| | - Soichi Yagi
- Department of Gastroenterology, Hyogo Medical University, Nishinomiya 663-8501, Japan
| | - Koji Kaku
- Department of Gastroenterology, Hyogo Medical University, Nishinomiya 663-8501, Japan
| | - Maiko Ikenouchi
- Department of Gastroenterology, Hyogo Medical University, Nishinomiya 663-8501, Japan
| | - Toshiyuki Sato
- Department of Gastroenterology, Hyogo Medical University, Nishinomiya 663-8501, Japan
| | - Koji Kamikozuru
- Department of Gastroenterology, Hyogo Medical University, Nishinomiya 663-8501, Japan
| | - Yoko Yokoyama
- Department of Gastroenterology, Hyogo Medical University, Nishinomiya 663-8501, Japan
| | - Tetsuya Takagawa
- Center for Clinical Research and Education, Hyogo Medical University, Nishinomiya 663-8501, Japan
| | - Masahito Shimizu
- Department of Gastroenterology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Shinichiro Shinzaki
- Department of Gastroenterology, Hyogo Medical University, Nishinomiya 663-8501, Japan
| |
Collapse
|
3
|
Cooney LN, O’Shea KD, Winfield HJ, Cahill MM, Pierce LT, McCarthy FO. Bisindolyl Maleimides and Indolylmaleimide Derivatives-A Review of Their Synthesis and Bioactivity. Pharmaceuticals (Basel) 2023; 16:1191. [PMID: 37764999 PMCID: PMC10534823 DOI: 10.3390/ph16091191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 09/29/2023] Open
Abstract
The evolution of bisindolyl maleimides and indolyl maleimide derivatives and their unique biological activities have stimulated great interest in medicinal chemistry programs. Bisindolylmaleimide (BIM)-type compounds arise from natural sources such as arcyriarubin and are biosynthetically related to indolocarbazoles. BIMs are commonly the immediate synthetic precursors of indolocarbazoles, lacking a central bond between the two aromatic units and making them more flexible and drug-like. Synthetic endeavours within this class of compounds are broad and have led to the development of both remarkably potent and selective protein kinase inhibitors. Clinical BIM examples include ruboxistaurin and enzastaurin, which are highly active inhibitors of protein kinase C-β. While BIMs are widely recognised as protein kinase inhibitors, other modes of activity have been reported, including the inhibition of calcium signalling and antimicrobial activity. Critically, structural differences can be used to exploit new bioactivity and therefore it is imperative to discover new chemical entities to address new targets. BIMs can be highly functionalised or chemically manipulated, which provides the opportunity to generate new derivatives with unique biological profiles. This review will collate new synthetic approaches to BIM-type compounds and their associated bioactivities with a focus on clinical applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Florence O. McCarthy
- School of Chemistry and ABCRF, University College Cork, Western Road, T12K8AF Cork, Ireland; (L.N.C.); (K.D.O.); (H.J.W.); (M.M.C.); (L.T.P.)
| |
Collapse
|
4
|
Agrawal A, Kulkarni GT. Topical application of aerial portion of Acalypha indica Linn ameliorates psoriasis in rodents: Evidences from in vivo and in silico studies. JOURNAL OF ETHNOPHARMACOLOGY 2023:116685. [PMID: 37236382 DOI: 10.1016/j.jep.2023.116685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/11/2023] [Accepted: 05/23/2023] [Indexed: 05/28/2023]
Abstract
ETHANOPHARMACOLOGICAL RELEVANCE Acalypha indica Linn. is a weed, used traditionally for different skin diseases such as eczema and dermatitis in various parts of India. There are no previous in vivo studies reported on the antipsoriatic potential of this medicinal plant. AIM The aim of this study was to investigate antipsoriatic activity of coconut oil dispersion of aerial portion of Acalypha indica Linn. Few lipid-soluble phytoconstituents of this plantwere subjected to molecular docking studies on different targets to determine phytoconstituent responsible for antipsoriatic activity. METHODS Virgin coconut oil dispersion of aerial portion of the plant was prepared by mixing three parts of coconut oil and one part of powdered aerial portion. The acute dermal toxicity was determined according to OECD guidelines. Mouse tail model was used to evaluate the antipsoriatic activity. Molecular docking of phytoconstituents was carried out using Biovia Discovery Studio. RESULTS In acute dermal toxicity study,the coconut oil dispersion was found to be safe up to the dose of 20000 mg/kg. The dispersion exhibited significant antipsoriatic activity (p < 0.01) at the dose of 250 mg/kg; at 500 mg/kg dose, the activity was similar that of 250 mg/kg dose. In the docking study of the phytoconstituents, 2-methyl anthraquinone was found to be responsible for antipsoriatic activity. CONCLUSION This study provides new evidence of Acalypha indica Linn as antipsoriatic plant and justifies its traditional use. Computational studies also endorse the results obtained via acute dermal toxicity study and mouse tail model for evaluation of antipsoriatic potential.
Collapse
Affiliation(s)
- Anurag Agrawal
- School of Pharmacy, ITM University, Gwalior, Madhya Pradesh, 474 001, India; Uttarakhand Technical University, Dehradun, Uttarakhand, 248 007, India; Department of Pharmacology, Ram-Eesh Institute of Vocational and Technical Education, Greater Noida, Dist. Gautam Buddha Nagar, Uttar Pradesh, 201310, India
| | - Giriraj T Kulkarni
- Gokaraju Rangaraju College of Pharmacy, Hyderabad, Telangana, 500 090, India.
| |
Collapse
|
5
|
Martins MS, Almeida IF, Cruz MT, Sousa E. Chronic pruritus: from pathophysiology to drug design. Biochem Pharmacol 2023; 212:115568. [PMID: 37116666 DOI: 10.1016/j.bcp.2023.115568] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 04/30/2023]
Abstract
Pruritus, the most common symptom in dermatology, is an innate response capable of protecting skin against irritants. Nonetheless, when it lasts more than six weeks it is assumed to be a chronic pathology having a negative impact on people's lives. Chronic pruritus (CP) can occur in common and rare skin diseases, having a high prevalence in global population. The existing therapies are unable to counteract CP or are associated with adverse effects, so the development of effective treatments is a pressing issue. The pathophysiological mechanisms underlying CP are not yet completely dissected but, based on current knowledge, involve a wide range of receptors, namely neurokinin 1 receptor (NK1R), Janus kinase (JAK), and transient receptor potential (TRP) ion channels, especially transient receptor potential vanilloid 1 (TRPV1) and transient receptor potential ankyrin 1 (TRPA1). This review will address the relevance of these molecular targets for the treatment of CP and molecules capable of modulating these receptors that have already been studied clinically or have the potential to possibly alleviate this pathology. According to scientific and clinical literature, there is an increase in the expression of these molecular targets in the lesioned skin of patients experiencing CP when compared with non-lesioned skin, highlighting their importance for the development of potential efficacious drugs through the design of antagonists/inhibitors.
Collapse
Affiliation(s)
- Márcia S Martins
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Novo Edifício do Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; Laboratory of Organic and Pharmaceutical Chemistry, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Isaobel F Almeida
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO-Applied Molecular Biosciences Unit, MedTech, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Maria T Cruz
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Emília Sousa
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Novo Edifício do Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; Laboratory of Organic and Pharmaceutical Chemistry, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
6
|
Atluri K, Manne S, Nalamothu V, Mantel A, Sharma PK, Babu RJ. Advances in Current Drugs and Formulations for the Management of Atopic Dermatitis. Crit Rev Ther Drug Carrier Syst 2023; 40:1-87. [PMID: 37585309 DOI: 10.1615/critrevtherdrugcarriersyst.2023042979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Atopic dermatitis (AD) is a chronic, relapsing inflammatory skin disease with a complex pathophysiology. Treatment of AD remains challenging owing to the presence of a wide spectrum of clinical phenotypes and limited response to existing therapies. However, recent genetic, immunological, and pathophysiological insights into the disease mechanism resulted in the invention of novel therapeutic drug candidates. This review provides a comprehensive overview of current therapies and assesses various novel drug delivery strategies currently under clinical investigation. Further, this review majorly emphasizes on various topical treatments including emollient therapies, barrier repair agents, topical corticosteroids (TCS), phosphodiesterase 4 (PDE4) inhibitors, calcineurin inhibitors, and Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway inhibitors. It also discusses biological and systemic therapies, upcoming treatments based on ongoing clinical trials. Additionally, this review scrutinized the use of pharmaceutical inactive ingredients in the approved topical dosage forms for AD treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - R Jayachandra Babu
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
7
|
Zhang Y, Xu Z, Zhan L, Gao Y, Zheng B, Zhou Y, Sheng Y, Liang G, Song Z. Design, synthesis and biological evaluation of novel chromone-maleimide hybrids as potent anti-inflammatory agents against LPS-induced acute lung injury. Bioorg Chem 2022; 128:106049. [DOI: 10.1016/j.bioorg.2022.106049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/06/2022] [Accepted: 07/20/2022] [Indexed: 11/02/2022]
|
8
|
Li S, Si H, Song X, Lei C, He X, Wang J, Liu Y, Zhou Y, Song JG, Peng L, Tang X, Chan S, Ren X, Tu Z, Li Z, Wang Z, Zhang Z, Ding K. Discovery of Hexahydrofuro[3,2- b]furans as New Kinase-Selective and Orally Bioavailable JAK3 Inhibitors for the Treatment of Leukemia Harboring a JAK3 Activating Mutant. J Med Chem 2022; 65:10674-10690. [PMID: 35860875 DOI: 10.1021/acs.jmedchem.2c00922] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Janus kinase 3 (JAK3) is a potential target for the treatment of hematological malignancies. Herein, we report the discovery of a series of new orally bioavailable irreversible JAK3 kinase inhibitors. The representative compound 12n potently inhibited JAK3 kinase activity with an IC50 value of 1.2 nM and was more than 900-fold selective over JAK1, JAK2, and Tyk2. Cell-based assays revealed that 12n significantly suppressed phosphorylation of JAK3 and the downstream effectors STAT3/5 and also robustly restrained proliferation of BaF3 cells transfected with JAK3M511I activating mutation and human leukemia U937 cells harboring JAK3M511I with IC50 values of 22.9 and 20.2 nM, respectively. More importantly, 12n showed reasonable pharmacokinetic (PK) properties, and oral administration of 12n at a dose of 50 mg/kg twice daily led to tumor regression in a U937 cell inoculated xenograft mouse model. Thus, 12n represents a promising lead compound for further optimization to discover new therapeutic agents for hematological malignancies.
Collapse
Affiliation(s)
- Shan Li
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Hongfei Si
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Xiaojuan Song
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Avenue, Guangzhou Science Park, Guangzhou 510530, China
| | - Chong Lei
- State Key Laboratory of Bioorganic Chemistry and Natural Products, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
| | - Xiaoqiang He
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Jie Wang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Yiling Liu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Yang Zhou
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Jian-Guo Song
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Lijie Peng
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Xia Tang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Shingpan Chan
- Guangzhou Lixin Pharmaceuticals, Guangzhou 510530, China
| | - Xiaomei Ren
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Zhengchao Tu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Avenue, Guangzhou Science Park, Guangzhou 510530, China
| | - Zhengqiu Li
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Zhen Wang
- State Key Laboratory of Bioorganic Chemistry and Natural Products, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
| | - Zhang Zhang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Ke Ding
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China.,State Key Laboratory of Bioorganic Chemistry and Natural Products, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China.,The First Affiliated Hospital (Huaqiao Hospital), Jinan University, #601 Huangpu Avenue West, Guangzhou 510632, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| |
Collapse
|
9
|
A Comprehensive Overview of Globally Approved JAK Inhibitors. Pharmaceutics 2022; 14:pharmaceutics14051001. [PMID: 35631587 PMCID: PMC9146299 DOI: 10.3390/pharmaceutics14051001] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/22/2022] [Accepted: 04/28/2022] [Indexed: 11/16/2022] Open
Abstract
Janus kinase (JAK) is a family of cytoplasmic non-receptor tyrosine kinases that includes four members, namely JAK1, JAK2, JAK3, and TYK2. The JAKs transduce cytokine signaling through the JAK-STAT pathway, which regulates the transcription of several genes involved in inflammatory, immune, and cancer conditions. Targeting the JAK family kinases with small-molecule inhibitors has proved to be effective in the treatment of different types of diseases. In the current review, eleven of the JAK inhibitors that received approval for clinical use have been discussed. These drugs are abrocitinib, baricitinib, delgocitinib, fedratinib, filgotinib, oclacitinib, pacritinib, peficitinib, ruxolitinib, tofacitinib, and upadacitinib. The aim of the current review was to provide an integrated overview of the chemical and pharmacological data of the globally approved JAK inhibitors. The synthetic routes of the eleven drugs were described. In addition, their inhibitory activities against different kinases and their pharmacological uses have also been explained. Moreover, their crystal structures with different kinases were summarized, with a primary focus on their binding modes and interactions. The proposed metabolic pathways and metabolites of these drugs were also illustrated. To sum up, the data in the current review could help in the design of new JAK inhibitors with potential therapeutic benefits in inflammatory and autoimmune diseases.
Collapse
|
10
|
Babu S, Nagarajan SK, Sathish S, Negi VS, Sohn H, Madhavan T. Identification of Potent and Selective JAK1 Lead Compounds Through Ligand-Based Drug Design Approaches. Front Pharmacol 2022; 13:837369. [PMID: 35529449 PMCID: PMC9068899 DOI: 10.3389/fphar.2022.837369] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/07/2022] [Indexed: 01/06/2023] Open
Abstract
JAK1 plays a significant role in the intracellular signaling by interacting with cytokine receptors in different types of cells and is linked to the pathogenesis of various cancers and in the pathology of the immune system. In this study, ligand-based pharmacophore modeling combined with virtual screening and molecular docking methods was incorporated to identify the potent and selective lead compounds for JAK1. Initially, the ligand-based pharmacophore models were generated using a set of 52 JAK1 inhibitors named C-2 methyl/hydroxyethyl imidazopyrrolopyridines derivatives. Twenty-seven pharmacophore models with five and six pharmacophore features were generated and validated using potency and selectivity validation methods. During potency validation, the Guner-Henry score was calculated to check the accuracy of the generated models, whereas in selectivity validation, the pharmacophore models that are capable of identifying selective JAK1 inhibitors were evaluated. Based on the validation results, the best pharmacophore models ADHRRR, DDHRRR, DDRRR, DPRRR, DHRRR, ADRRR, DDHRR, and ADPRR were selected and taken for virtual screening against the Maybridge, Asinex, Chemdiv, Enamine, Lifechemicals, and Zinc database to identify the new molecules with novel scaffold that can bind to JAK1. A total of 4,265 hits were identified from screening and checked for acceptable drug-like properties. A total of 2,856 hits were selected after ADME predictions and taken for Glide molecular docking to assess the accurate binding modes of the lead candidates. Ninety molecules were shortlisted based on binding energy and H-bond interactions with the important residues of JAK1. The docking results were authenticated by calculating binding free energy for protein–ligand complexes using the MM-GBSA calculation and induced fit docking methods. Subsequently, the cross-docking approach was carried out to recognize the selective JAK1 lead compounds. Finally, top five lead compounds that were potent and selective against JAK1 were selected and validated using molecular dynamics simulation. Besides, the density functional theory study was also carried out for the selected leads. Through various computational studies, we observed good potency and selectivity of these lead compounds when compared with the drug ruxolitinib. Compounds such as T5923555 and T5923531 were found to be the best and can be further validated using in vitro and in vivo methods.
Collapse
Affiliation(s)
- Sathya Babu
- Computational Biology Lab, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, India
| | - Santhosh Kumar Nagarajan
- Computational Biology Lab, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, India
| | - Sruthy Sathish
- Computational Biology Lab, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, India
| | - Vir Singh Negi
- Department of Clinical Immunology, Jawaharlal Institute of Post-Graduate Medical Education and Research, Pondicherry, India
| | - Honglae Sohn
- Department of Chemistry and Department of Carbon Materials, Chosun University, Gwangju, South Korea
- *Correspondence: Thirumurthy Madhavan, ; Honglae Sohn,
| | - Thirumurthy Madhavan
- Computational Biology Lab, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, India
- *Correspondence: Thirumurthy Madhavan, ; Honglae Sohn,
| |
Collapse
|
11
|
Vargas DF, Kaufman TS, Larghi EL. Total Synthesis of Aqabamycin G, a Nitrophenyl Indolylmaleimide Marine Alkaloid from Vibrio sp. WMBA. J Org Chem 2022; 87:13494-13500. [PMID: 35324169 DOI: 10.1021/acs.joc.2c00063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The first total synthesis of the marine alkaloid aqabamycin G is disclosed. The synthetic sequence involved the stepwise addition to maleimide of an indole motif and a substituted diazo-benzenoid unit derived from acetaminophen. An alternative strategy using a protected phenol is also reported.
Collapse
Affiliation(s)
- Didier F Vargas
- Instituto de Química Rosario (IQUIR, CONICET-UNR) and Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, S2002LRK Rosario, Argentina
| | - Teodoro S Kaufman
- Instituto de Química Rosario (IQUIR, CONICET-UNR) and Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, S2002LRK Rosario, Argentina
| | - Enrique L Larghi
- Instituto de Química Rosario (IQUIR, CONICET-UNR) and Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, S2002LRK Rosario, Argentina
| |
Collapse
|
12
|
Murer L, Volle R, Andriasyan V, Petkidis A, Gomez-Gonzalez A, Yang L, Meili N, Suomalainen M, Bauer M, Policarpo Sequeira D, Olszewski D, Georgi F, Kuttler F, Turcatti G, Greber UF. Identification of broad anti-coronavirus chemical agents for repurposing against SARS-CoV-2 and variants of concern. CURRENT RESEARCH IN VIROLOGICAL SCIENCE 2022; 3:100019. [PMID: 35072124 PMCID: PMC8760634 DOI: 10.1016/j.crviro.2022.100019] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/04/2022] [Accepted: 01/04/2022] [Indexed: 01/18/2023]
Abstract
Endemic human coronaviruses (hCoVs) 229E and OC43 cause respiratory disease with recurrent infections, while severe acute respiratory syndrome (SARS)-CoV-2 spreads across the world with impact on health and societies. Here, we report an image-based multicycle infection procedure with α-coronavirus hCoV-229E-eGFP in an arrayed chemical library screen of 5440 clinical and preclinical compounds. Toxicity counter selection and challenge with the β-coronaviruses OC43 and SARS-CoV-2 in tissue culture and human airway epithelial explant cultures (HAEEC) identified four FDA-approved compounds with oral availability. Methylene blue (MB, used for the treatment of methemoglobinemia), Mycophenolic acid (MPA, used in organ transplantation) and the anti-fungal agent Posaconazole (POS) had the broadest anti-CoV spectrum. They inhibited the shedding of SARS-CoV-2 and variants-of-concern (alpha, beta, gamma, delta) from HAEEC in either pre- or post exposure regimens at clinically relevant concentrations. Co-treatment of cultured cells with MB and the FDA-approved SARS-CoV-2 RNA-polymerase inhibitor Remdesivir reduced the effective anti-viral concentrations of MB by 2-fold, and Remdesivir by 4 to 10-fold, indicated by BLISS independence synergy modelling. Neither MB, nor MPA, nor POS affected the cell delivery of SARS-CoV-2 or OC43 (+)sense RNA, but blocked subsequent viral RNA accumulation in cells. Unlike Remdesivir, MB, MPA or POS did not reduce the release of viral RNA in post exposure regimen, thus indicating infection inhibition at a post-replicating step as well. In summary, the data emphasize the power of unbiased, full cycle compound screens to identify and repurpose broadly acting drugs against coronaviruses.
Collapse
Affiliation(s)
- Luca Murer
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Romain Volle
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Vardan Andriasyan
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Anthony Petkidis
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Alfonso Gomez-Gonzalez
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Liliane Yang
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Nicole Meili
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Maarit Suomalainen
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Michael Bauer
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Daniela Policarpo Sequeira
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Dominik Olszewski
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Fanny Georgi
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Fabien Kuttler
- Biomolecular Screening Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Station 15, 1015, Lausanne, Switzerland
| | - Gerardo Turcatti
- Biomolecular Screening Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Station 15, 1015, Lausanne, Switzerland
| | - Urs F Greber
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| |
Collapse
|
13
|
Zhou S, Huang G. Some important inhibitors and mechanisms of rheumatoid arthritis. Chem Biol Drug Des 2021; 99:930-943. [PMID: 34942050 DOI: 10.1111/cbdd.14015] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 11/29/2022]
Abstract
Rheumatoid arthritis is a chronic disease that seriously affects human health and quality of life, and it is one of the main causes of labor loss and disability. Many countries have listed rheumatoid arthritis as one of the national a key diseases to tackle. The pathogenesis of RA in humans is still unknown, and medical researchers believe that the pathogenesis of RA may be the result of a combination of genetic and environmental factors. RA is an incurable condition that can only be controlled and treated with conventional drugs. In this paper, the pathologic features and pathogenesis of RA were introduced, and the research progress of new anti-rheumatoid arthritis chemical drugs in recent years was reviewed.
Collapse
Affiliation(s)
- Shiyang Zhou
- Chongqing Chemical Industry Vocational College, Chongqing, 401228, China.,College of Chemistry, Chongqing Normal University, Chongqing, 401331, China
| | - Gangliang Huang
- College of Chemistry, Chongqing Normal University, Chongqing, 401331, China
| |
Collapse
|
14
|
Zhang N, Zhang C, Zeng Z, Zhang J, Du S, Bao C, Wang Z. Preclinical Characterization of the Selective JAK1 Inhibitor LW402 for Treatment of Rheumatoid Arthritis. J Inflamm Res 2021; 14:2133-2147. [PMID: 34054304 PMCID: PMC8153205 DOI: 10.2147/jir.s301076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/01/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Research on JAK family members as therapeutic targets for autoimmune diseases has brought tofacitinib and baricitinib into clinical for the treatment of rheumatoid arthritis and other autoimmune diseases. Despite the potent efficacy of these first-generation JAK inhibitors, their broad-spectrum JAK inhibition and adverse events warrant development of a JAK1-specific inhibitor to improve their safety profile. METHODS In this study, we characterized a JAK1-specific inhibitor, LW402, on biochemical and human whole-blood assays. We further evaluated the therapeutic efficacy of LW402 in a rat adjuvant-induced arthritis (rAIA) model and a mouse collagen-induced arthritis (mCIA) model. The safety of LW402 was evaluated in both SpragueDawley rats and cynomolgus monkeys. RESULTS LW402 exhibited potent nanomolar activity against JAK1 and showed a 45-fold selectivity for inhibition of JAK1- over JAK2-dependent signaling induced by either IL6 or GM-CSF in human whole-blood assays. In the rAIA model, oral dosing of LW402 resulted in a dose-dependent improvement in disease symptoms, including reduction in paw swelling, marked reduction in the inflammatory-cell infiltration to synovial tissue, and protection of articular cartilage and bone from damage. The therapeutic efficacy of LW402 correlated well with the plasma exposure of LW402 and the extent of pSTAT3 inhibition in white blood cells. LW402 also effectively eased disease symptoms in the mCIA model. Toxicity studies in the Sprague Dawley rats and cynomolgus monkeys established a ≥5x therapeutic window for LW402 as drug exposures of toxicity study NOAEL dose and pharmacology study ED50 dose were compared. CONCLUSION We developed a novel JAK1-specific inhibitor LW402 with potent efficacy in rAIA and mCIA models. We established a good safety profile for LW402 in toxicity studies, and the overall superiority of LW402 should translated well to the clinical setting for the treatment of RA and other autoimmune diseases.
Collapse
Affiliation(s)
- Ning Zhang
- Longwood Biopharmaceuticals, Shanghai, People’s Republic of China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Zhihong Zeng
- Longwood Biopharmaceuticals, Shanghai, People’s Republic of China
| | - Jiyong Zhang
- Longwood Biopharmaceuticals, Shanghai, People’s Republic of China
| | - Shengnan Du
- Longwood Biopharmaceuticals, Shanghai, People’s Republic of China
| | - Chunde Bao
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Zhe Wang
- Longwood Biopharmaceuticals, Shanghai, People’s Republic of China
| |
Collapse
|
15
|
Remenyi J, Naik RJ, Wang J, Razsolkov M, Verano A, Cai Q, Tan L, Toth R, Raggett S, Baillie C, Traynor R, Hastie CJ, Gray NS, Arthur JSC. Generation of a chemical genetic model for JAK3. Sci Rep 2021; 11:10093. [PMID: 33980892 PMCID: PMC8115619 DOI: 10.1038/s41598-021-89356-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 03/26/2021] [Indexed: 01/17/2023] Open
Abstract
Janus Kinases (JAKs) have emerged as an important drug target for the treatment of a number of immune disorders due to the central role that they play in cytokine signalling. 4 isoforms of JAKs exist in mammalian cells and the ideal isoform profile of a JAK inhibitor has been the subject of much debate. JAK3 has been proposed as an ideal target due to its expression being largely restricted to the immune system and its requirement for signalling by cytokine receptors using the common γ-chain. Unlike other JAKs, JAK3 possesses a cysteine in its ATP binding pocket and this has allowed the design of isoform selective covalent JAK3 inhibitors targeting this residue. We report here that mutating this cysteine to serine does not prevent JAK3 catalytic activity but does greatly increase the IC50 for covalent JAK3 inhibitors. Mice with a Cys905Ser knockin mutation in the endogenous JAK3 gene are viable and show no apparent welfare issues. Cells from these mice show normal STAT phosphorylation in response to JAK3 dependent cytokines but are resistant to the effects of covalent JAK3 inhibitors. These mice therefore provide a chemical-genetic model to study JAK3 function.
Collapse
Affiliation(s)
- Judit Remenyi
- Division of Cell Signalling and Immunology, School of Life Sciences, Wellcome Trust Building, University of Dundee, Dundee, DD1 5EH, UK
| | - Rangeetha Jayaprakash Naik
- Division of Cell Signalling and Immunology, School of Life Sciences, Wellcome Trust Building, University of Dundee, Dundee, DD1 5EH, UK
| | - Jinhua Wang
- Department of Cancer Biology, Dana Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, USA
| | - Momchil Razsolkov
- Division of Cell Signalling and Immunology, School of Life Sciences, Wellcome Trust Building, University of Dundee, Dundee, DD1 5EH, UK
| | - Alyssa Verano
- Department of Cancer Biology, Dana Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, USA
| | - Quan Cai
- Department of Cancer Biology, Dana Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, USA
| | - Li Tan
- Department of Cancer Biology, Dana Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, USA
| | - Rachel Toth
- MRC PPU Reagents and Services, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Samantha Raggett
- MRC PPU Reagents and Services, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Carla Baillie
- MRC PPU Reagents and Services, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Ryan Traynor
- MRC PPU Reagents and Services, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - C James Hastie
- MRC PPU Reagents and Services, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Nathanael S Gray
- Department of Cancer Biology, Dana Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, USA
| | - J Simon C Arthur
- Division of Cell Signalling and Immunology, School of Life Sciences, Wellcome Trust Building, University of Dundee, Dundee, DD1 5EH, UK.
| |
Collapse
|
16
|
Yu J, Rao L, Zhan L, Wang B, Zhan Q, Xu Y, Zhao H, Wang X, Zhou Y, Guo Y, Wu X, Song Z, Yu F. The small molecule ZY-214-4 may reduce the virulence of Staphylococcus aureus by inhibiting pigment production. BMC Microbiol 2021; 21:67. [PMID: 33639851 PMCID: PMC7916275 DOI: 10.1186/s12866-021-02113-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/02/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND In recent years, clinical Staphylococcus aureus isolates have become highly resistant to antibiotics, which has raised concerns about the ability to control infections by these organisms. The aim of this study was to clarify the effect of a new small molecule, ZY-214-4 (C19H11BrNO4), on S. aureus pigment production. RESULTS At the concentration of 4 μg/mL, ZY-214-4 exerted a significant inhibitory effect on S. aureus pigment synthesis, without affecting its growth or inducing a toxic effect on the silkworm. An oxidant sensitivity test and a whole-blood killing test indicated that the S. aureus survival rate decreased significantly with ZY-214-4 treatment. Additionally, ZY-214-4 administration significantly reduced the expression of a pigment synthesis-related gene (crtM) and the superoxide dismutase genes (sodA) as determined by real-time quantitative polymerase chain reaction (RT-qPCR) analysis. ZY-214-4 treatment also improved the survival rate of S. aureus-infected silkworm larvae. CONCLUSIONS The small molecule ZY-214-4 has potential for the prevention of S. aureus infections by reducing the virulence associated with this bacterium.
Collapse
Affiliation(s)
- Jingyi Yu
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Lulin Rao
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Lingling Zhan
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Bingjie Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200082, China
| | - Qing Zhan
- Nanchang University, Nanchang, 330027, China
| | - Yanlei Xu
- Nanchang University, Nanchang, 330027, China
| | - Huilin Zhao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200082, China
| | - Xinyi Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200082, China
| | - Yan Zhou
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yinjuan Guo
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200082, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200082, China
| | - Xiaocui Wu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200082, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200082, China
| | - Zengqiang Song
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, China.
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200082, China.
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200082, China.
| |
Collapse
|
17
|
Yu J, Rao L, Zhan L, Zhou Y, Guo Y, Wu X, Song Z, Yu F. Antibiofilm Activity of Small-Molecule ZY-214-4 Against Staphylococcus aureus. Front Microbiol 2021; 12:618922. [PMID: 33613488 PMCID: PMC7886693 DOI: 10.3389/fmicb.2021.618922] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/11/2021] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus is the most important pathogenic bacteria in humans. As the resistance of S. aureus to existing antibiotics is increasing, there is an urgent need for new anti-infective drugs. S. aureus biofilms cause persistent infections and resist complete eradication with antibiotic therapy. The present study investigated the inhibitory effect of the novel small-molecule ZY-214-4 (C19H11BrNO4) on S. aureus biofilm formation. At a subinhibitory concentration (4 μg/ml), ZY-214-4 had no effect on the growth of S. aureus strains and also showed no cytotoxicity in human normal bronchial epithelial cells (Bease-2B). The results of a semi-quantitative biofilm test showed that ZY-214-4 prevented S. aureus biofilm formation, which was confirmed by scanning electron microscopy and confocal laser scanning microscopy. ZY-214-4 significantly suppressed the production of polysaccharide intercellular adhesion and prevented cell aggregation, and also inhibited the mRNA expression of icaA and other biofilm-related genes (eno, clfA/B, fnbB, fib, ebpS, psmα, and psmβ) in clinical S. aureus isolates. Thus, at a subinhibitory concentration, ZY-214-4 inhibits biofilm formation by preventing cell aggregation, highlighting its clinical potential for preventing or treating S. aureus infections.
Collapse
Affiliation(s)
- Jingyi Yu
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lulin Rao
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lingling Zhan
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Zhou
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yinjuan Guo
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaocui Wu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zengqiang Song
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Ge T, Jhala G, Fynch S, Akazawa S, Litwak S, Pappas EG, Catterall T, Vakil I, Long AJ, Olson LM, Krishnamurthy B, Kay TW, Thomas HE. The JAK1 Selective Inhibitor ABT 317 Blocks Signaling Through Interferon-γ and Common γ Chain Cytokine Receptors to Reverse Autoimmune Diabetes in NOD Mice. Front Immunol 2020; 11:588543. [PMID: 33343569 PMCID: PMC7746546 DOI: 10.3389/fimmu.2020.588543] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/04/2020] [Indexed: 01/31/2023] Open
Abstract
Cytokines that signal through the JAK-STAT pathway, such as interferon-γ (IFN-γ) and common γ chain cytokines, contribute to the destruction of insulin-secreting β cells by CD8+ T cells in type 1 diabetes (T1D). We previously showed that JAK1/JAK2 inhibitors reversed autoimmune insulitis in non-obese diabetic (NOD) mice and also blocked IFN-γ mediated MHC class I upregulation on β cells. Blocking interferons on their own does not prevent diabetes in knockout NOD mice, so we tested whether JAK inhibitor action on signaling downstream of common γ chain cytokines, including IL-2, IL-7 IL-15, and IL-21, may also affect the progression of diabetes in NOD mice. Common γ chain cytokines activate JAK1 and JAK3 to regulate T cell proliferation. We used a JAK1-selective inhibitor, ABT 317, to better understand the specific role of JAK1 signaling in autoimmune diabetes. ABT 317 reduced IL-21, IL-2, IL-15 and IL-7 signaling in T cells and IFN-γ signaling in β cells, but ABT 317 did not affect GM-CSF signaling in granulocytes. When given in vivo to NOD mice, ABT 317 reduced CD8+ T cell proliferation as well as the number of KLRG+ effector and CD44hiCD62Llo effector memory CD8+ T cells in spleen. ABT 317 also prevented MHC class I upregulation on β cells. Newly diagnosed diabetes was reversed in 94% NOD mice treated twice daily with ABT 317 while still on treatment at 40 days and 44% remained normoglycemic after a further 60 days from discontinuing the drug. Our results indicate that ABT 317 blocks common γ chain cytokines in lymphocytes and interferons in lymphocytes and β cells and are thus more effective against diabetes pathogenesis than IFN-γ receptor deficiency alone. Our studies suggest use of this class of drug for the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Tingting Ge
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia.,Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | - Gaurang Jhala
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
| | - Stacey Fynch
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
| | - Satoru Akazawa
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
| | - Sara Litwak
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
| | - Evan G Pappas
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
| | - Tara Catterall
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
| | - Ishan Vakil
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia.,Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | - Andrew J Long
- AbbVie Bioresearch Center, Worcester, MA, United States
| | - Lisa M Olson
- AbbVie Bioresearch Center, Worcester, MA, United States
| | - Balasubramanian Krishnamurthy
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia.,Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | - Thomas W Kay
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia.,Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | - Helen E Thomas
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia.,Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| |
Collapse
|
19
|
Fernández-Clotet A, Castro-Poceiro J, Panés J. JAK Inhibition: The Most Promising Agents in the IBD Pipeline? Curr Pharm Des 2019; 25:32-40. [DOI: 10.2174/1381612825666190405141410] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 03/01/2019] [Indexed: 12/12/2022]
Abstract
Under current therapeutic algorithms, half of the patients with moderate-severe ulcerative colitis or
Crohn’s disease fail in achieving a sustained remission. New drugs with different mechanisms of action are
needed. After two decades of new drug avenues in inflammatory bowel disease dominated by the development of
monoclonal antibodies, in recent years we are witnessing promising developments of small molecules for these
conditions. Their intrinsic characteristics make them attractive compared to the monoclonal antibodies based on
their oral administration, short plasma half-life, lack of immunogenicity and predictable pharmacokinetics.
Among them, Janus kinase (JAK) inhibitors are a promising new class that have demonstrated efficacy with a
favorable safety profile in clinical trials. Tofacitinib has been the first JAK inhibitor approved for the treatment of
ulcerative colitis. This review discusses the molecular aspects of the JAK-STAT pathway, its role in the pathogenesis
of inflammatory bowel disease, and the rational use of JAK inhibitors in these conditions. The different
compounds with JAK inhibitory activity tested are reviewed and we provide an overview of recent evidence from
clinical trials. Finally, we consider the positioning of these drugs in the treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Agnès Fernández-Clotet
- Inflammatory Bowel Disease Group, Institut d'Investigacions Biometiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Jesús Castro-Poceiro
- Inflammatory Bowel Disease Group, Institut d'Investigacions Biometiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Julián Panés
- Inflammatory Bowel Disease Group, Institut d'Investigacions Biometiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
20
|
Are peptides a solution for the treatment of hyperactivated JAK3 pathways? Inflammopharmacology 2019; 27:433-452. [PMID: 30929155 DOI: 10.1007/s10787-019-00589-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/18/2019] [Indexed: 01/10/2023]
Abstract
While the inactivation mutations that eliminate JAK3 function lead to the immunological disorders such as severe combined immunodeficiency, activation mutations, causing constitutive JAK3 signaling, are known to trigger various types of cancer or are responsible for autoimmune diseases, such as rheumatoid arthritis, psoriasis, or inflammatory bowel diseases. Treatment of hyperactivated JAK3 is still an obstacle, due to different sensibility of mutation types to conventional drugs and unwanted side effects, because these drugs are not absolutely specific for JAK3, thus inhibiting other members of the JAK family, too. Lack of information, in which way sole inhibition of JAK3 is necessary for elimination of the disease, calls for the development of isoform-specific JAK3 inhibitors. Beside this strategy, up to date peptides are a rising alternative as chemo- or immunotherapeutics, but still sparsely represented in drug development and clinical trials. Beyond a possible direct inhibition function, crossing the cancer cell membrane and interfering in disease-causing pathways or triggering apoptosis, peptides could be used in future as adjunct remedies to potentialize traditional therapy and preserve non-affected cells. To discuss such feasible topics, this review deals with the knowledge about the structure-function of JAK3 and the actual state-of-the-art of isoform-specific inhibitor development, as well as the function of currently approved drugs or those currently being tested in clinical trials. Furthermore, several strategies for the application of peptide-based drugs for cancer therapy and the physicochemical and structural relations to peptide efficacy are discussed, and an overview of peptide sequences, which were qualified for clinical trials, is given.
Collapse
|
21
|
Shi L, Zhong Z, Li X, Zhou Y, Pan Z. Discovery of an Orally Available Janus Kinase 3 Selective Covalent Inhibitor. J Med Chem 2019; 62:1054-1066. [PMID: 30615446 DOI: 10.1021/acs.jmedchem.8b01823] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
JAK family kinases are important mediators of immune cell signaling and Janus Kinase 3 (JAK3) has long been indicated as a potential target for autoimmune disorders. Intensive efforts to develop highly selective JAK3 inhibitors have been underway for many years. However, because of JAK3's strong binding preference to adenosine 5'-triphosphate (ATP), a number of inhibitors exhibit large gaps between enzymatic and cellular potency, which hampers efforts to dissect the roles of JAK3 in cellular settings. Using a targeted covalent inhibitor approach, we discovered compound 32, which overcame ATP competition (1 mM) in the enzymatic assay, and demonstrated significantly improved inhibitory activity for JAK3-dependent signaling in mouse CTLL-2 and human peripheral blood mononuclear cells. Compound 32 also exhibited high selectivity within the JAK family and good pharmacokinetic properties. Thus, it may serve as a highly valuable tool molecule to study the overlapping roles of JAK family kinases in complex biological settings. Our study also suggested that for covalent kinase inhibitors, especially those targeting kinases with low Km ATP values, the reversible interactions between molecules and proteins should be carefully optimized to improve the overall potency.
Collapse
Affiliation(s)
- Liyang Shi
- State Key Laboratory of Chemical Oncogenomics, Engineering Laboratory for Chiral Drug Synthesis, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School , Peking University , Shenzhen 518055 , China
| | - Zhenpeng Zhong
- State Key Laboratory of Chemical Oncogenomics, Engineering Laboratory for Chiral Drug Synthesis, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School , Peking University , Shenzhen 518055 , China
| | - Xitao Li
- State Key Laboratory of Chemical Oncogenomics, Engineering Laboratory for Chiral Drug Synthesis, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School , Peking University , Shenzhen 518055 , China
| | - Yiqing Zhou
- State Key Laboratory of Chemical Oncogenomics, Engineering Laboratory for Chiral Drug Synthesis, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School , Peking University , Shenzhen 518055 , China
| | - Zhengying Pan
- State Key Laboratory of Chemical Oncogenomics, Engineering Laboratory for Chiral Drug Synthesis, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School , Peking University , Shenzhen 518055 , China
| |
Collapse
|
22
|
Other Forms of Immunosuppression. KIDNEY TRANSPLANTATION - PRINCIPLES AND PRACTICE 2019. [PMCID: PMC7152196 DOI: 10.1016/b978-0-323-53186-3.00020-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
23
|
Casimiro-Garcia A, Trujillo JI, Vajdos F, Juba B, Banker ME, Aulabaugh A, Balbo P, Bauman J, Chrencik J, Coe JW, Czerwinski R, Dowty M, Knafels JD, Kwon S, Leung L, Liang S, Robinson RP, Telliez JB, Unwalla R, Yang X, Thorarensen A. Identification of Cyanamide-Based Janus Kinase 3 (JAK3) Covalent Inhibitors. J Med Chem 2018; 61:10665-10699. [DOI: 10.1021/acs.jmedchem.8b01308] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | - John I. Trujillo
- Medicine Design, Pfizer Inc., 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Felix Vajdos
- Medicine Design, Pfizer Inc., 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | | | - Mary Ellen Banker
- Medicine Design, Pfizer Inc., 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Ann Aulabaugh
- Medicine Design, Pfizer Inc., 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | | | - Jonathan Bauman
- Medicine Design, Pfizer Inc., 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jill Chrencik
- Medicine Design, Pfizer Inc., 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jotham W. Coe
- Medicine Design, Pfizer Inc., 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | | | - Martin Dowty
- Medicine Design, Pfizer Inc., 1 Burtt Road, Andover, Massachusetts 01810, United States
| | - John D. Knafels
- Medicine Design, Pfizer Inc., 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Soojin Kwon
- Medicine Design, Pfizer Inc., 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Louis Leung
- Medicine Design, Pfizer Inc., 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Sidney Liang
- Medicine Design, Pfizer Inc., 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Ralph P. Robinson
- Medicine Design, Pfizer Inc., 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | | | | | - Xin Yang
- Medicine Design, Pfizer Inc., 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | | |
Collapse
|
24
|
Jasuja H, Chadha N, Singh PK, Kaur M, Bahia MS, Silakari O. Putative dual inhibitors of Janus kinase 1 and 3 (JAK1/3): Pharmacophore based hierarchical virtual screening. Comput Biol Chem 2018; 76:109-117. [PMID: 29990790 DOI: 10.1016/j.compbiolchem.2018.07.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 04/01/2018] [Accepted: 07/04/2018] [Indexed: 11/26/2022]
Abstract
Janus kinase 1 and 3 are non-receptor protein tyrosine kinases, involved in the regulation of various cytokines implicated in the pathogenesis of autoimmune and inflammatory disease conditions. Thus, they serve as therapeutic targets for the designing of multi-targeted agents for the treatment of inflammatory-mediated pathological conditions. In the present study, diverse inhibitors of JAK1 and JAK3 were considered for the development of ligand-based pharmacophore models, followed by docking analysis to design putative dual inhibitors. The pharmacophore models were generated in PHASE 3.4, and top five models for each target were selected on the basis of survival minus inactive score. The best model for JAK1 (AAADH.25) and JAK3 (ADDRR.142) were selected corresponding to the highest value of Q2test. Both models were employed for the screening of a PHASE database, and subsequently, the retrieved hits were filtered employing molecular docking in JAK1 and JAK3 proteins. The stable interactions between retrieved hits and proteins were confirmed using molecular dynamics simulations. Finally, ADME properties of screened dual inhibitors displaying essential interactions with both proteins were calculated. Thus, the new leads obtained in this way may be prioritized for experimental validation as potential novel therapeutic agents in the treatment of various autoimmune and inflammatory disorders related to JAK1 and JAK3.
Collapse
Affiliation(s)
- Haneesh Jasuja
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Navriti Chadha
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Pankaj Kumar Singh
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Maninder Kaur
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Malkeet Singh Bahia
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Om Silakari
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India.
| |
Collapse
|
25
|
Forster M, Chaikuad A, Dimitrov T, Döring E, Holstein J, Berger BT, Gehringer M, Ghoreschi K, Müller S, Knapp S, Laufer SA. Development, Optimization, and Structure-Activity Relationships of Covalent-Reversible JAK3 Inhibitors Based on a Tricyclic Imidazo[5,4- d]pyrrolo[2,3- b]pyridine Scaffold. J Med Chem 2018; 61:5350-5366. [PMID: 29852068 DOI: 10.1021/acs.jmedchem.8b00571] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Janus kinases are major drivers of immune signaling and have been the focus of anti-inflammatory drug discovery for more than a decade. Because of the invariable colocalization of JAK1 and JAK3 at cytokine receptors, the question if selective JAK3 inhibition is sufficient to effectively block downstream signaling has been highly controversial. Recently, we discovered the covalent-reversible JAK3 inhibitor FM-381 (23) featuring high isoform and kinome selectivity. Crystallography revealed that this inhibitor induces an unprecedented binding pocket by interactions of a nitrile substituent with arginine residues in JAK3. Herein, we describe detailed structure-activity relationships necessary for induction of the arginine pocket and the impact of this structural change on potency, isoform selectivity, and efficacy in cellular models. Furthermore, we evaluated the stability of this novel inhibitor class in in vitro metabolic assays and were able to demonstrate an adequate stability of key compound 23 for in vivo use.
Collapse
Affiliation(s)
- Michael Forster
- Department of Pharmaceutical/Medicinal Chemistry , Eberhard Karls University Tübingen , Auf der Morgenstelle 8 , 72076 Tübingen , DE , Germany
| | - Apirat Chaikuad
- Institute for Pharmaceutical Chemistry , Johann Wolfgang Goethe University , Max-von-Laue-Str. 9 , D-60438 Frankfurt am Main , DE , Germany.,Structural Genomics Consortium , Johann Wolfgang Goethe University , Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Str. 15 , D-60438 Frankfurt am Main , DE , Germany
| | - Teodor Dimitrov
- Department of Pharmaceutical/Medicinal Chemistry , Eberhard Karls University Tübingen , Auf der Morgenstelle 8 , 72076 Tübingen , DE , Germany
| | - Eva Döring
- Department of Pharmaceutical/Medicinal Chemistry , Eberhard Karls University Tübingen , Auf der Morgenstelle 8 , 72076 Tübingen , DE , Germany
| | - Julia Holstein
- Department of Dermatology , University Medical Center, Eberhard Karls University Tübingen , Liebermeisterstr. 25 , 72076 Tübingen , DE , Germany
| | - Benedict-Tilman Berger
- Institute for Pharmaceutical Chemistry , Johann Wolfgang Goethe University , Max-von-Laue-Str. 9 , D-60438 Frankfurt am Main , DE , Germany.,Structural Genomics Consortium , Johann Wolfgang Goethe University , Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Str. 15 , D-60438 Frankfurt am Main , DE , Germany
| | - Matthias Gehringer
- Department of Pharmaceutical/Medicinal Chemistry , Eberhard Karls University Tübingen , Auf der Morgenstelle 8 , 72076 Tübingen , DE , Germany
| | - Kamran Ghoreschi
- Department of Dermatology , University Medical Center, Eberhard Karls University Tübingen , Liebermeisterstr. 25 , 72076 Tübingen , DE , Germany
| | - Susanne Müller
- Institute for Pharmaceutical Chemistry , Johann Wolfgang Goethe University , Max-von-Laue-Str. 9 , D-60438 Frankfurt am Main , DE , Germany
| | - Stefan Knapp
- Institute for Pharmaceutical Chemistry , Johann Wolfgang Goethe University , Max-von-Laue-Str. 9 , D-60438 Frankfurt am Main , DE , Germany.,Structural Genomics Consortium , Johann Wolfgang Goethe University , Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Str. 15 , D-60438 Frankfurt am Main , DE , Germany.,German Cancer Consortium, DKTK , 60590 Site Frankfurt/Mainz , DE , Germany
| | - Stefan A Laufer
- Department of Pharmaceutical/Medicinal Chemistry , Eberhard Karls University Tübingen , Auf der Morgenstelle 8 , 72076 Tübingen , DE , Germany
| |
Collapse
|
26
|
O'Mahony A, John MR, Cho H, Hashizume M, Choy EH. Discriminating phenotypic signatures identified for tocilizumab, adalimumab, and tofacitinib monotherapy and their combinations with methotrexate. J Transl Med 2018; 16:156. [PMID: 29879987 PMCID: PMC5992722 DOI: 10.1186/s12967-018-1532-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/30/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Clinical trials have shown combinations of anti-tumor necrosis factor biologicals plus methotrexate (MTX) are more effective treatments for rheumatoid arthritis than biological monotherapies, based, in part, on the assumption that MTX reduces the immunogenicity of biologicals. However, co-treatment with the anti-interleukin-6 receptor-alpha antibody tocilizumab (TCZ) and MTX does not demonstrate the same level of incremental benefit over TCZ monotherapy. Using the human primary cell based BioMAP phenotypic profiling platform, we investigated the impact of TCZ, adalimumab (ADA), and the small molecule drug tofacitinib (TOF), alone and in combination with MTX, on translational biomarkers that could indicate unique pharmacodynamic interactions outside those of reduced immunogenicity. METHODS TCZ, ADA, and TOF, alone and in combination with MTX, were profiled in BioMAP systems at concentrations close to clinical exposure levels: TCZ, 200 μg/ml; TOF1, 1.1 μM; TOF2, 0.12 µM; MTX, 10 μM. Changes in biomarkers were evaluated by statistical methods to determine whether combinations differed from the individual agents. RESULTS Although the BioMAP activity profile for TCZ + MTX was not significantly different from that for TCZ alone, profiles for ADA + MTX and TOF1 + MTX or TOF2 + MTX had a greater number of statistically significant different activities (P < 0.01) than did agents profiled individually. CONCLUSIONS These data support the comparable efficacy of TCZ as monotherapy and as combination therapy and suggest that TOF, like ADA, may be more beneficial in combination with MTX. Taking an orthogonal approach to directly compare monotherapy and combination therapies indicates that MTX contributes to the efficacy of some, but not all, RA therapies and can be affected by factors additional to reduced immunogenicity.
Collapse
Affiliation(s)
- Alison O'Mahony
- BioMAP Division, Eurofins DiscoverX, 310 Utah Avenue, South San Francisco, CA, 94080, USA.
| | | | - Hannah Cho
- BioMAP Division, Eurofins DiscoverX, 310 Utah Avenue, South San Francisco, CA, 94080, USA
| | | | - Ernest H Choy
- Division of Infection and Immunity, CREATE Centre, Cardiff University, Cardiff, CF10 3AT, UK
| |
Collapse
|
27
|
Development of selective inhibitors for the treatment of rheumatoid arthritis: (R)-3-(3-(Methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)pyrrolidin-1-yl)-3-oxopropanenitrile as a JAK1-selective inhibitor. Bioorg Med Chem 2018; 26:1495-1510. [DOI: 10.1016/j.bmc.2018.01.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/22/2018] [Accepted: 01/24/2018] [Indexed: 01/01/2023]
|
28
|
Pei H, He L, Shao M, Yang Z, Ran Y, Li D, Zhou Y, Tang M, Wang T, Gong Y, Chen X, Yang S, Xiang M, Chen L. Discovery of a highly selective JAK3 inhibitor for the treatment of rheumatoid arthritis. Sci Rep 2018; 8:5273. [PMID: 29588471 PMCID: PMC5869712 DOI: 10.1038/s41598-018-23569-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 03/15/2018] [Indexed: 02/05/2023] Open
Abstract
Janus tyrosine kinase 3 (JAK3) is expressed in lymphoid cells and is involved in the signalling of T cell functions. The development of a selective JAK3 inhibitor has been shown to have a potential benefit in the treatment of autoimmune disorders. In this article, we developed the 4-aminopiperidine-based compound RB1, which was highly selective for JAK3 inhibition, with an IC50 of value of 40 nM, but did not inhibit JAK1, JAK2 or tyrosine kinase 2 (TYK2) at concentrations up to 5 µM. Furthermore, RB1 also exhibited favourable selectivity against a panel of representative kinases. In a battery of cytokine-stimulated cell-based assays, this potent inhibitor of JAK3 activity with good selectivity against other kinases could potently inhibit JAK3 activity over the activity of JAK1 or JAK2 (over at least 100-fold). A combination of liquid chromatography-mass spectrometry (LC-MS) experiments validated that RB1 covalently modified the unique cysteine 909 residue in JAK3. In vivo, RB1 exerted significantly improved pathology in the joints of a collagen-induced arthritis mouse model. The reasonable pharmacokinetics properties (F = 72.52%, T1/2 = 14.6 h) and favourable results of toxicology experiments (LD50 > 2 g/kg) suggest that RB1 has the potential to be an efficacious treatment for RA.
Collapse
Affiliation(s)
- Heying Pei
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Linhong He
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Mingfeng Shao
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Zhuang Yang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yan Ran
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Dan Li
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yuanyuan Zhou
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Minghai Tang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Taijin Wang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yanqiu Gong
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Xiaoxin Chen
- Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan, Guangdong, 523325, China
| | - Shengyong Yang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Mingli Xiang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, China. .,Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan, Guangdong, 523325, China.
| | - Lijuan Chen
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, China. .,Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan, Guangdong, 523325, China.
| |
Collapse
|
29
|
Fowler NJ, Blanford CF, de Visser SP, Warwicker J. Features of reactive cysteines discovered through computation: from kinase inhibition to enrichment around protein degrons. Sci Rep 2017; 7:16338. [PMID: 29180682 PMCID: PMC5703995 DOI: 10.1038/s41598-017-15997-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/26/2017] [Indexed: 02/07/2023] Open
Abstract
Large-scale characterisation of cysteine modification is enabling study of the physicochemical determinants of reactivity. We find that location of cysteine at the amino terminus of an α-helix, associated with activity in thioredoxins, is under-represented in human protein structures, perhaps indicative of selection against background reactivity. An amino-terminal helix location underpins the covalent linkage for one class of kinase inhibitors. Cysteine targets for S-palmitoylation, S-glutathionylation, and S-nitrosylation show little correlation with pKa values predicted from structures, although flanking sequences of S-palmitoylated sites are enriched in positively-charged amino acids, which could facilitate palmitoyl group transfer to substrate cysteine. A surprisingly large fraction of modified sites, across the three modifications, would be buried in native protein structure. Furthermore, modified cysteines are (on average) closer to lysine ubiquitinations than are unmodified cysteines, indicating that cysteine redox biology could be associated with protein degradation and degron recognition.
Collapse
Affiliation(s)
- Nicholas J Fowler
- The Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, United Kingdom.,School of Chemistry, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, United Kingdom
| | - Christopher F Blanford
- The Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, United Kingdom.,School of Materials, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, United Kingdom
| | - Sam P de Visser
- The Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, United Kingdom.,School of Chemical Engineering and Analytical Science, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, United Kingdom
| | - Jim Warwicker
- The Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, United Kingdom. .,School of Chemistry, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, United Kingdom.
| |
Collapse
|
30
|
Molecular dynamics and integrated pharmacophore-based identification of dual [Formula: see text] inhibitors. Mol Divers 2017; 22:95-112. [PMID: 29138965 DOI: 10.1007/s11030-017-9794-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 10/24/2017] [Indexed: 12/14/2022]
Abstract
Despite increase in the understanding of the pathogenesis of rheumatoid arthritis (RA), it remains a tough challenge. The advent of kinases involved in key intracellular pathways in pathogenesis of RA may provide a new phase of drug discovery for RA. The present study is aimed to identify dual JAK3/[Formula: see text] inhibitors by developing an optimum pharmacophore model integrating the information revealed by ligand-based pharmacophore models and structure-based pharmacophore models (SBPMs). For JAK3 inhibitors, the addition of an aromatic ring feature and for [Formula: see text] the addition of a hydrophobic feature proposed by SBPMs lead to five-point pharmacophore (i.e., AADHR.54 (JAK3)) and six-point pharmacophore (i.e., AAAHRR.45 ([Formula: see text])). The obtained pharmacophores were validated and used for virtual screening and then for docking-based screening. Molecules were further evaluated for ADME properties, and their docked protein complexes were subjected to MM-GBSA energy calculations and molecular dynamic simulations. The top two hit compounds with novel scaffolds 2-oxo-1,2-dihydroquinoline and benzo[d]oxazole showed inhibitory activity for JAK3 and [Formula: see text].
Collapse
|
31
|
Forster M, Gehringer M, Laufer SA. Recent advances in JAK3 inhibition: Isoform selectivity by covalent cysteine targeting. Bioorg Med Chem Lett 2017; 27:4229-4237. [PMID: 28844493 DOI: 10.1016/j.bmcl.2017.07.079] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 07/27/2017] [Accepted: 07/28/2017] [Indexed: 01/01/2023]
Abstract
Janus kinases (JAKs) are a family of four cytosolic protein kinases with a high degree of structural similarity. Due to its very restricted role in immune regulation, JAK3 was promoted as an excellent target for immunosuppression for more than a decade, but clinical validation of this concept is still elusive. During the last years, speculation arose that kinase activity of JAK1, which cooperates with JAK3 in cytokine receptor signaling, may have a dominant role over the one of JAK3. Until recently, however, this issue could not be appropriately addressed due to a lack of highly isoform-selective tool compounds. With the recent resurgence of covalent drugs, targeting of a specific cysteine that distinguishes JAK3 from other JAK family members became an attractive design option. By applying this strategy, a set of JAK3 inhibitors with excellent selectivity against other JAK isoforms and the kinome was developed during the last three years and used to decipher JAK3-dependent signaling. The data obtained with these tool compounds demonstrates that selective JAK3 inhibition is sufficient to block downstream signaling. Since one of these inhibitors is currently under evaluation in phase II clinical studies against several inflammatory disorders, it will soon become apparent whether selective JAK3 inhibition translates into clinical efficacy.
Collapse
Affiliation(s)
- Michael Forster
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard-Karls-University Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Matthias Gehringer
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard-Karls-University Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany.
| | - Stefan A Laufer
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard-Karls-University Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany.
| |
Collapse
|
32
|
Oxindole-based SYK and JAK3 dual inhibitors for rheumatoid arthritis: designing, synthesis and biological evaluation. Future Med Chem 2017; 9:1193-1211. [DOI: 10.4155/fmc-2017-0037] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: Autoimmune disorders have complex pathophysiology and focus is laid on the development of multitargeted agents. Two well-established kinases: SYK and JAK3, were considered to design dual inhibitors as potential therapeutics using various molecular-modeling approaches. Mehodology: Pharmacophore models for SYK and JAK3 were generated using oxindole-based inhibitors. Furthermore, an in-house database was designed that was screened against the best selected models. The obtained hits were employed for docking analysis and subjected to MM-GBSA analysis and molecular dynamic simulation. Results: Top five oxindole derivatives were synthesized and evaluated for in vitro SYK and JAK3 activity. The most active compound 4a was evaluated for in vivo antiarthritic activity. It showed significant anti-arthritic activity. Conclusion: Thus, the designed inhibitors resulted in potential therapeutic agents for rheumatoid arthritis.
Collapse
|
33
|
Seif F, Khoshmirsafa M, Aazami H, Mohsenzadegan M, Sedighi G, Bahar M. The role of JAK-STAT signaling pathway and its regulators in the fate of T helper cells. Cell Commun Signal 2017. [PMID: 28637459 PMCID: PMC5480189 DOI: 10.1186/s12964-017-0177-y] [Citation(s) in RCA: 533] [Impact Index Per Article: 66.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway plays critical roles in orchestrating of immune system, especially cytokine receptors and they can modulate the polarization of T helper cells. This pathway is regulated by an array of regulator proteins, including Suppressors of Cytokine Signaling (SOCS), Protein Inhibitors of Activated STATs (PIAS) and Protein Tyrosine Phosphatases (PTPs) determining the initiation, duration and termination of the signaling cascades. Dysregulation of the JAK-STAT pathway in T helper cells may result in various immune disorders. In this review, we represent how the JAK-STAT pathway is generally regulated and then in Th cell subsets in more detail. Finally, we introduce novel targeted strategies as promising therapeutic approaches in the treatment of immune disorders. Studies are ongoing for identifying the other regulators of the JAK-STAT pathway and designing innovative therapeutic strategies. Therefore, further investigation is needed.
Collapse
Affiliation(s)
- Farhad Seif
- ENT and Head and Neck Research Center and Department, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran.,Department of immunology, school of medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Khoshmirsafa
- Department of immunology, school of medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Aazami
- Department of immunology, school of medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Monireh Mohsenzadegan
- Department of Medical Laboratory Science, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Sedighi
- Department of immunology, school of medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadali Bahar
- Department of immunology, school of medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
The Selective JAK1/3-Inhibitor R507 Mitigates Obliterative Airway Disease Both With Systemic Administration and Aerosol Inhalation. Transplantation 2017; 100:1022-31. [PMID: 26910327 DOI: 10.1097/tp.0000000000001110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND The efficacy of selective Janus kinase 1/3 inhibitor R507 to prevent obliterative airway disease was analyzed in preclinical airway transplantation models. METHODS Orthotopic trachea transplantations were performed between Lewis donors and Brown Norway rat recipients. Oral everolimus (4 mg/kg once per day) or oral respective inhaled R507 (60 mg/kg twice per day, each) was used for immunosuppression. Grafts were retrieved after 6 or 60 days. Toxicity and anti-inflammatory effects of R507 were analyzed on human airway epithelial cells. RESULTS In 6-day animals, oral and inhaled R507 more potently diminished mononuclear graft infiltration than everolimus and preserved ciliated pseudostratified columnar respiratory epithelium. Everolimus and R507 similarly suppressed systemic cellular and humoral immune activation. In untreated rats, marked obliterative airway disease developed over 60 days. Oral and inhaled R507 was significantly more effective in reducing airway obliteration and preserved the morphology of the airway epithelium. Luciferase-positive donors revealed that a substantial amount of smooth muscle cells within the obliterative tissue was of donor origin. Only everolimus but not R507, adversely altered kidney function and lipid profiles. The R507 aerosol did not show airway toxicity in vitro but effectively suppressed activation of inflammatory signaling pathways induced by IL-1β. CONCLUSIONS The Janus kinase 1/3 inhibitor R507 is a very well-tolerated immunosuppressant that similarly diminished obliterative airway disease with systemic or inhaled administration.
Collapse
|
35
|
Elwood F, Witter DJ, Piesvaux J, Kraybill B, Bays N, Alpert C, Goldenblatt P, Qu Y, Ivanovska I, Lee HH, Chiu CS, Tang H, Scott ME, Deshmukh SV, Zielstorff M, Byford A, Chakravarthy K, Dorosh L, Rivkin A, Klappenbach J, Pan BS, Kariv I, Dinsmore C, Slipetz D, Dandliker PJ. Evaluation of JAK3 Biology in Autoimmune Disease Using a Highly Selective, Irreversible JAK3 Inhibitor. J Pharmacol Exp Ther 2017; 361:229-244. [PMID: 28193636 DOI: 10.1124/jpet.116.239723] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/03/2017] [Indexed: 03/08/2025] Open
Abstract
Reversible janus associated kinase (JAK) inhibitors such as tofacitinib and decernotinib block cytokine signaling and are efficacious in treating autoimmune diseases. However, therapeutic doses are limited due to inhibition of other JAK/signal transducer and activator of transcription pathways associated with hematopoiesis, lipid biogenesis, infection, and immune responses. A selective JAK3 inhibitor may have a better therapeutic index; however, until recently, no compounds have been described that maintain JAK3 selectivity in cells, as well as against the kinome, with good physicochemical properties to test the JAK3 hypothesis in vivo. To quantify the biochemical basis for JAK isozyme selectivity, we determined that the apparent Km value for each JAK isozyme ranged from 31.8 to 2.9 μM for JAK1 and JAK3, respectively. To confirm compound activity in cells, we developed a novel enzyme complementation assay that read activity of single JAK isozymes in a cellular context. Reversible JAK3 inhibitors cannot achieve sufficient selectivity against other isozymes in the cellular context due to inherent differences in enzyme ATP Km values. Therefore, we developed irreversible JAK3 compounds that are potent and highly selective in vitro in cells and against the kinome. Compound 2, a potent inhibitor of JAK3 (0.15 nM) was 4300-fold selective for JAK3 over JAK1 in enzyme assays, 67-fold [interleukin (IL)-2 versus IL-6] or 140-fold [IL-2 versus erythropoietin or granulocyte-macrophage colony-stimulating factor (GMCSF)] selective in cellular reporter assays and >35-fold selective in human peripheral blood mononuclear cell assays (IL-7 versus IL-6 or GMCSF). In vivo, selective JAK3 inhibition was sufficient to block the development of inflammation in a rat model of rheumatoid arthritis, while sparing hematopoiesis.
Collapse
Affiliation(s)
- Fiona Elwood
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - David J Witter
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Jennifer Piesvaux
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Brian Kraybill
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Nathan Bays
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Carla Alpert
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Peter Goldenblatt
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Yujie Qu
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Irena Ivanovska
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Hyun-Hee Lee
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Chi-Sung Chiu
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Hao Tang
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Mark E Scott
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Sujal V Deshmukh
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Mark Zielstorff
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Alan Byford
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Kalyan Chakravarthy
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Lauren Dorosh
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Alexey Rivkin
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Joel Klappenbach
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Bo-Sheng Pan
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Ilona Kariv
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Christopher Dinsmore
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Deborah Slipetz
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| | - Peter J Dandliker
- Departments of Respiratory and Immunology (F.E., Y.Q., I.I., H.-H.L., H.T., D.S.), Chemistry (D.W., M.E.S., A.R., C.D.), Pharmacology (J.P., B.K., N.B., C.A., P.G., M.Z., A.B., K.C., B.-S.P., I.K., P.J.D.), and Pharmacokinetics, Pharmacodynamics and Drug Metabolism (C.-S.C., S.V.D., L.D., J.K.), Merck Research Laboratories, Boston, Massachusetts
| |
Collapse
|
36
|
Abstract
Tofacitinib is the first Janus kinase (JAK) inhibitor commercially approved for the treatment of rheumatoid arthritis. This compound and a number of other JAK inhibitors are currently being tested in phase II and III trials for the treatment of a variety of autoimmune inflammatory diseases. Whereas a characteristic safety profile is emerging for some JAK inhibitors, differences between individual agents might emerge on the basis of distinct potency against their molecular targets. Similarly to biological therapy, JAK inhibition can lead to serious and opportunistic infections, and viral infections seem to be particularly frequent. Although no malignancy signals have been identified to date, long-term follow-up and further research are needed to understand the risk of malignancy associated with these compounds. As is the case for biologic agents, vaccination is important to mitigate the risks of these emerging therapies.
Collapse
Affiliation(s)
- Kevin L Winthrop
- Oregon Health &Science University, GH104, 3375 SW Terwilliger Blvd. Portland, Oregon, 97239, USA
| |
Collapse
|
37
|
Moore CA, Iasella CJ, Venkataramanan R, Lakkis FG, Smith RB, McDyer JF, Zeevi A, Ensor CR. Janus kinase inhibition for immunosuppression in solid organ transplantation: Is there a role in complex immunologic challenges? Hum Immunol 2016; 78:64-71. [PMID: 27998802 DOI: 10.1016/j.humimm.2016.12.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/14/2016] [Accepted: 12/15/2016] [Indexed: 01/02/2023]
Abstract
Inhibition of the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway for immunosuppression in solid organ transplantation is appealing due to its specificity for immune cell function, particularly for JAK3. This is due to its unique association with only the common gamma chain (γc). The γc is an appealing immunosuppression target in transplantation because of the critically important lymphokines that act at it, including IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. Tofacitinib was initially purported to selectively inhibit solely JAK3, but subsequent analyses have also demonstrated its activity at the other members of the JAK family. Clinical outcomes have validated tofacitinib's pan-JAK activity in kidney transplantation after demonstrating an increased risk of infection and malignancy as compared to CNI-based regimens. After these trials, tofacitinib investigation for use in transplantation has effectively ceased. However, a post-hoc analysis has shed new light on the monitoring of tofacitinib exposure in order to predict infection and oncologic events. With new methods to monitor tofacitinib exposure, clinicians may be able to effectively reduce toxicities while providing a high level of immunosuppression. The purpose of this review to identify when, and for whom, JAK inhibitors may provide benefit in solid organ transplantation.
Collapse
Affiliation(s)
- Cody A Moore
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States.
| | - Carlo J Iasella
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Raman Venkataramanan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States; Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Fadi G Lakkis
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Thomas E. Starzl Transplantation Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Randall B Smith
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - John F McDyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Adriana Zeevi
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Christopher R Ensor
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States; Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
38
|
Kaur M, Silakari O. Ligand-based and e-pharmacophore modeling, 3D-QSAR and hierarchical virtual screening to identify dual inhibitors of spleen tyrosine kinase (Syk) and janus kinase 3 (JAK3). J Biomol Struct Dyn 2016; 35:3043-3060. [PMID: 27678281 DOI: 10.1080/07391102.2016.1240108] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The clinical efficacy of multiple kinase inhibitors has caught the interest of Pharmaceutical and Biotech researchers to develop potential drugs with multi-kinase inhibitory activity for complex diseases. In the present work, we attempted to identify dual inhibitors of spleen tyrosine kinase (Syk) and janus kinase 3 (JAK3), keys players in immune signaling, by developing ideal pharmacophores integrating Ligand-based pharmacophore models (LBPMs) and Structure-based pharmacophore models (SBPMs), thereby projecting the optimum pharmacophoric required for inhibition of both the kinases. The four point LBPM; ADPR.14 suggested the presence of one hydrogen bond acceptor, one hydrogen bond donor, one positive ionizable, and one ring aromatic feature for Syk inhibitory activity and AADH.54 proposed the necessity of two hydrogen bond acceptor, one hydrogen bond donor, and one hydrophobic feature for JAK3 inhibitory activity. To our interest, SBPMs identified additional ring aromatic features required for inhibition of both the kinases. For Syk inhibitory activity, the hydrogen bond acceptor feature indicated by LBPM was devoid of forming hydrogen bonding interaction with the hinge region amino acid residue (Ala451). Thus merging the information revealed by both LBPMs and SBPMs, ideal pharmacophore models i.e. ADPRR.14 (Syk) and AADHR.54 (JAK3) were generated. These models after rigorous statistical validation were used for screening of Asinex database. The systematic virtual screening protocol, including pharmacophore and docking-based screening, ADME property, and MM-GBSA energy calculations, retrieved final 10 hits as dual inhibitors of Syk and JAK3. Final 10 hits thus obtained can aid in the development of potential therapeutic agents for autoimmune disorders. Also the top two hits were evaluated against both the enzymes.
Collapse
Affiliation(s)
- Maninder Kaur
- a Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research , Punjabi University , Patiala , Punjab 147002 , India
| | - Om Silakari
- a Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research , Punjabi University , Patiala , Punjab 147002 , India
| |
Collapse
|
39
|
Forster M, Chaikuad A, Bauer SM, Holstein J, Robers MB, Corona CR, Gehringer M, Pfaffenrot E, Ghoreschi K, Knapp S, Laufer SA. Selective JAK3 Inhibitors with a Covalent Reversible Binding Mode Targeting a New Induced Fit Binding Pocket. Cell Chem Biol 2016; 23:1335-1340. [PMID: 27840070 PMCID: PMC5119931 DOI: 10.1016/j.chembiol.2016.10.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/21/2016] [Accepted: 10/13/2016] [Indexed: 01/01/2023]
Abstract
Janus kinases (JAKs) are a family of cytoplasmatic tyrosine kinases that are attractive targets for the development of anti-inflammatory drugs given their roles in cytokine signaling. One question regarding JAKs and their inhibitors that remains under intensive debate is whether JAK inhibitors should be isoform selective. Since JAK3 functions are restricted to immune cells, an isoform-selective inhibitor for JAK3 could be especially valuable to achieve clinically more useful and precise effects. However, the high degree of structural conservation makes isoform-selective targeting a challenging task. Here, we present picomolar inhibitors with unprecedented kinome-wide selectivity for JAK3. Selectivity was achieved by concurrent covalent reversible targeting of a JAK3-specific cysteine residue and a ligand-induced binding pocket. We confirmed that in vitro activity and selectivity translate well into the cellular environment and suggest that our inhibitors are powerful tools to elucidate JAK3-specific functions.
Identification and characterization of novel covalent reversible JAK3 inhibitors Picomolar affinities along with both high isoform and kinome selectivity is achieved Covalent-reversible interaction and a new induced binding pocket confirmed by X-ray structures High potency and selectivity are successfully proven in cellular models
Collapse
Affiliation(s)
- Michael Forster
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard-Karls-University Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Apirat Chaikuad
- Nuffield Department of Clinical Medicine, Structural Genomics Consortium and Target Discovery Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Silke M Bauer
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard-Karls-University Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Julia Holstein
- Department of Dermatology, University Medical Center, Eberhard-Karls-University Tuebingen, Liebermeisterstraße 25, 72076 Tuebingen, Germany
| | - Matthew B Robers
- Promega Corporation, 2800 Woods Hollow Road, Madison, WI 53711, USA
| | - Cesear R Corona
- Promega Corporation, 2800 Woods Hollow Road, Madison, WI 53711, USA
| | - Matthias Gehringer
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard-Karls-University Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Ellen Pfaffenrot
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard-Karls-University Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Kamran Ghoreschi
- Department of Dermatology, University Medical Center, Eberhard-Karls-University Tuebingen, Liebermeisterstraße 25, 72076 Tuebingen, Germany
| | - Stefan Knapp
- Nuffield Department of Clinical Medicine, Structural Genomics Consortium and Target Discovery Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK.
| | - Stefan A Laufer
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard-Karls-University Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany.
| |
Collapse
|
40
|
Bajusz D, Ferenczy GG, Keserű GM. Discovery of Subtype Selective Janus Kinase (JAK) Inhibitors by Structure-Based Virtual Screening. J Chem Inf Model 2015; 56:234-47. [DOI: 10.1021/acs.jcim.5b00634] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Dávid Bajusz
- Medicinal Chemistry Research
Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., Budapest 1117, Hungary
| | - György G. Ferenczy
- Medicinal Chemistry Research
Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., Budapest 1117, Hungary
| | - György M. Keserű
- Medicinal Chemistry Research
Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., Budapest 1117, Hungary
| |
Collapse
|
41
|
Losdyck E, Hornakova T, Springuel L, Degryse S, Gielen O, Cools J, Constantinescu SN, Flex E, Tartaglia M, Renauld JC, Knoops L. Distinct Acute Lymphoblastic Leukemia (ALL)-associated Janus Kinase 3 (JAK3) Mutants Exhibit Different Cytokine-Receptor Requirements and JAK Inhibitor Specificities. J Biol Chem 2015; 290:29022-34. [PMID: 26446793 DOI: 10.1074/jbc.m115.670224] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Indexed: 01/22/2023] Open
Abstract
JAK1 and JAK3 are recurrently mutated in acute lymphoblastic leukemia. These tyrosine kinases associate with heterodimeric cytokine receptors such as IL-7 receptor or IL-9 receptor, in which JAK1 is appended to the specific chain, and JAK3 is appended to the common gamma chain. Here, we studied the role of these receptor complexes in mediating the oncogenic activity of JAK3 mutants. Although JAK3(V674A) and the majority of other JAK3 mutants needed to bind to a functional cytokine receptor complex to constitutively activate STAT5, JAK3(L857P) was unexpectedly found to not depend on such receptor complexes for its activity, which was induced without receptor or JAK1 co-expression. Introducing a mutation in the FERM domain that abolished JAK-receptor interaction did not affect JAK3(L857P) activity, whereas it inhibited the other receptor-dependent mutants. The same cytokine receptor independence as for JAK3(L857P) was observed for homologous Leu(857) mutations of JAK1 and JAK2 and for JAK3(L875H). This different cytokine receptor requirement correlated with different functional properties in vivo and with distinct sensitivity to JAK inhibitors. Transduction of murine hematopoietic cells with JAK3(V674A) led homogenously to lymphoblastic leukemias in BALB/c mice. In contrast, transduction with JAK3(L857P) induced various types of lymphoid and myeloid leukemias. Moreover, ruxolitinib, which preferentially blocks JAK1 and JAK2, abolished the proliferation of cells transformed by the receptor-dependent JAK3(V674A), yet proved much less potent on cells expressing JAK3(L857P). These particular cells were, in contrast, more sensitive to JAK3-specific inhibitors. Altogether, our results showed that different JAK3 mutations induce constitutive activation through distinct mechanisms, pointing to specific therapeutic perspectives.
Collapse
Affiliation(s)
- Elisabeth Losdyck
- From the Ludwig Institute for Cancer Research, Brussels Branch and the de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Tekla Hornakova
- From the Ludwig Institute for Cancer Research, Brussels Branch and the de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Lorraine Springuel
- From the Ludwig Institute for Cancer Research, Brussels Branch and the de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Sandrine Degryse
- the VIB Center for the Biology of Disease, K.U. Leuven, 3000 Leuven, Belgium, the K.U. Leuven Center for Human Genetics, K.U. Leuven, 3000 Leuven, Belgium
| | - Olga Gielen
- the VIB Center for the Biology of Disease, K.U. Leuven, 3000 Leuven, Belgium, the K.U. Leuven Center for Human Genetics, K.U. Leuven, 3000 Leuven, Belgium
| | - Jan Cools
- the VIB Center for the Biology of Disease, K.U. Leuven, 3000 Leuven, Belgium, the K.U. Leuven Center for Human Genetics, K.U. Leuven, 3000 Leuven, Belgium
| | - Stefan N Constantinescu
- From the Ludwig Institute for Cancer Research, Brussels Branch and the de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | | | - Marco Tartaglia
- the Genetic Disorders and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesu' IRCCS, Viale di San Paolo 15, 00146 Rome, Italy
| | - Jean-Christophe Renauld
- From the Ludwig Institute for Cancer Research, Brussels Branch and the de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Laurent Knoops
- From the Ludwig Institute for Cancer Research, Brussels Branch and the de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium, the Hematology Unit, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium, and
| |
Collapse
|
42
|
Farmer LJ, Ledeboer MW, Hoock T, Arnost MJ, Bethiel RS, Bennani YL, Black JJ, Brummel CL, Chakilam A, Dorsch WA, Fan B, Cochran JE, Halas S, Harrington EM, Hogan JK, Howe D, Huang H, Jacobs DH, Laitinen LM, Liao S, Mahajan S, Marone V, Martinez-Botella G, McCarthy P, Messersmith D, Namchuk M, Oh L, Penney MS, Pierce AC, Raybuck SA, Rugg A, Salituro FG, Saxena K, Shannon D, Shlyakter D, Swenson L, Tian SK, Town C, Wang J, Wang T, Wannamaker MW, Winquist RJ, Zuccola HJ. Discovery of VX-509 (Decernotinib): A Potent and Selective Janus Kinase 3 Inhibitor for the Treatment of Autoimmune Diseases. J Med Chem 2015; 58:7195-216. [DOI: 10.1021/acs.jmedchem.5b00301] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Luc J. Farmer
- Vertex Pharmaceuticals (Canada) Inc., 275 Armand-Frappier, Laval, Québec H7V 4A7, Canada
| | - Mark W. Ledeboer
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Thomas Hoock
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Michael J. Arnost
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Randy S. Bethiel
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Youssef L. Bennani
- Vertex Pharmaceuticals (Canada) Inc., 275 Armand-Frappier, Laval, Québec H7V 4A7, Canada
| | - James J. Black
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Christopher L. Brummel
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | | | - Warren A. Dorsch
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Bin Fan
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - John E. Cochran
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Summer Halas
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Edmund M. Harrington
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - James K. Hogan
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - David Howe
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Hui Huang
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Dylan H. Jacobs
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Leena M. Laitinen
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Shengkai Liao
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Sudipta Mahajan
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Valerie Marone
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | | | - Pamela McCarthy
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - David Messersmith
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Mark Namchuk
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Luke Oh
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Marina S. Penney
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Albert C. Pierce
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Scott A. Raybuck
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Arthur Rugg
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Francesco G. Salituro
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Kumkum Saxena
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Dean Shannon
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Dina Shlyakter
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Lora Swenson
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Shi-Kai Tian
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Christopher Town
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Jian Wang
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Tiansheng Wang
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - M. Woods Wannamaker
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Raymond J. Winquist
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Harmon J. Zuccola
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| |
Collapse
|
43
|
Tan L, Akahane K, McNally R, Reyskens KMSE, Ficarro SB, Liu S, Herter-Sprie GS, Koyama S, Pattison MJ, Labella K, Johannessen L, Akbay EA, Wong KK, Frank DA, Marto JA, Look TA, Arthur JSC, Eck MJ, Gray NS. Development of Selective Covalent Janus Kinase 3 Inhibitors. J Med Chem 2015; 58:6589-606. [PMID: 26258521 DOI: 10.1021/acs.jmedchem.5b00710] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The Janus kinases (JAKs) and their downstream effectors, signal transducer and activator of transcription proteins (STATs), form a critical immune cell signaling circuit, which is of fundamental importance in innate immunity, inflammation, and hematopoiesis, and dysregulation is frequently observed in immune disease and cancer. The high degree of structural conservation of the JAK ATP binding pockets has posed a considerable challenge to medicinal chemists seeking to develop highly selective inhibitors as pharmacological probes and as clinical drugs. Here we report the discovery and optimization of 2,4-substituted pyrimidines as covalent JAK3 inhibitors that exploit a unique cysteine (Cys909) residue in JAK3. Investigation of structure-activity relationship (SAR) utilizing biochemical and transformed Ba/F3 cellular assays resulted in identification of potent and selective inhibitors such as compounds 9 and 45. A 2.9 Å cocrystal structure of JAK3 in complex with 9 confirms the covalent interaction. Compound 9 exhibited decent pharmacokinetic properties and is suitable for use in vivo. These inhibitors provide a set of useful tools to pharmacologically interrogate JAK3-dependent biology.
Collapse
Affiliation(s)
- Li Tan
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School , Boston, Massachusetts 02115, United States
| | | | - Randall McNally
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School , Boston, Massachusetts 02115, United States
| | - Kathleen M S E Reyskens
- Division of Cell Signaling and Immunology, College of Life Sciences, University of Dundee , Dundee DD1 5EH. U.K
| | - Scott B Ficarro
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School , Boston, Massachusetts 02115, United States
| | | | | | | | - Michael J Pattison
- Division of Cell Signaling and Immunology, College of Life Sciences, University of Dundee , Dundee DD1 5EH. U.K
| | | | - Liv Johannessen
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School , Boston, Massachusetts 02115, United States
| | | | | | | | - Jarrod A Marto
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School , Boston, Massachusetts 02115, United States
| | | | - J Simon C Arthur
- Division of Cell Signaling and Immunology, College of Life Sciences, University of Dundee , Dundee DD1 5EH. U.K
| | - Michael J Eck
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School , Boston, Massachusetts 02115, United States
| | - Nathanael S Gray
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School , Boston, Massachusetts 02115, United States
| |
Collapse
|
44
|
Nakajima Y, Inoue T, Nakai K, Mukoyoshi K, Hamaguchi H, Hatanaka K, Sasaki H, Tanaka A, Takahashi F, Kunikawa S, Usuda H, Moritomo A, Higashi Y, Inami M, Shirakami S. Synthesis and evaluation of novel 1H-pyrrolo[2,3-b]pyridine-5-carboxamide derivatives as potent and orally efficacious immunomodulators targeting JAK3. Bioorg Med Chem 2015; 23:4871-4883. [DOI: 10.1016/j.bmc.2015.05.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/14/2015] [Accepted: 05/15/2015] [Indexed: 01/09/2023]
|
45
|
Zimmermann K, Sang X, Mastalerz HA, Johnson WL, Zhang G, Liu Q, Batt D, Lombardo LJ, Vyas D, Trainor GL, Tokarski JS, Lorenzi MV, You D, Gottardis MM, Lippy J, Khan J, Sack JS, Purandare AV. 9H-Carbazole-1-carboxamides as potent and selective JAK2 inhibitors. Bioorg Med Chem Lett 2015; 25:2809-12. [PMID: 25987372 DOI: 10.1016/j.bmcl.2015.04.101] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 04/28/2015] [Accepted: 04/30/2015] [Indexed: 02/07/2023]
Abstract
The discovery, synthesis, and characterization of 9H-carbazole-1-carboxamides as potent and selective ATP-competitive inhibitors of Janus kinase 2 (JAK2) are discussed. Optimization for JAK family selectivity led to compounds 14 and 21, with greater than 45-fold selectivity for JAK2 over all other members of the JAK kinase family.
Collapse
Affiliation(s)
- Kurt Zimmermann
- Bristol-Myers Squibb Co., 5 Research Parkway, Wallingford, CT 06492-1951, USA.
| | - Xiaopeng Sang
- Bristol-Myers Squibb Co., 5 Research Parkway, Wallingford, CT 06492-1951, USA
| | - Harold A Mastalerz
- Bristol-Myers Squibb Co., 5 Research Parkway, Wallingford, CT 06492-1951, USA
| | - Walter L Johnson
- Bristol-Myers Squibb Co., 5 Research Parkway, Wallingford, CT 06492-1951, USA
| | - Guifen Zhang
- Bristol-Myers Squibb Co., 5 Research Parkway, Wallingford, CT 06492-1951, USA
| | - Qingjie Liu
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Douglas Batt
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Louis J Lombardo
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Dinesh Vyas
- Bristol-Myers Squibb Co., 5 Research Parkway, Wallingford, CT 06492-1951, USA
| | - George L Trainor
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - John S Tokarski
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Matthew V Lorenzi
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Dan You
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Marco M Gottardis
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Jonathan Lippy
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Javed Khan
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - John S Sack
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Ashok V Purandare
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| |
Collapse
|
46
|
Ubel C, Mousset S, Trufa D, Sirbu H, Finotto S. Establishing the role of tyrosine kinase 2 in cancer. Oncoimmunology 2014; 2:e22840. [PMID: 23482926 PMCID: PMC3583936 DOI: 10.4161/onci.22840] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Tyrosine kinase 2 (TYK2) is a member of the Janus family of non-receptor tyrosine kinases involved in cytokine signaling. TYK2 deficiency is associated with increased susceptibility to mycobacterial and viral infections, hyper IgE syndrome as well as with allergic asthma. However the precise role of TYK2 in oncogenesis and tumor progression is not clear yet. Tyk2-deficient mice are prone to develop tumors because they lack efficient cytotoxic CD8+ T-cell antitumor responses as a result of deficient Type I interferon signaling. However, as TYK2 functions downstream of growth factor receptors that are often hyperactivated in cancer, inhibiting TYK2 might also have beneficial effects for cancer treatment.
Collapse
Affiliation(s)
- Caroline Ubel
- Laboratory of Cellular and Molecular Lung Immunology; Institute of Molecular Pneumology; University of Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | |
Collapse
|
47
|
Higashi Y. [JAK inhibitors as a new generation of small-molecule immunosuppressants]. Nihon Yakurigaku Zasshi 2014; 144:160-166. [PMID: 25312284 DOI: 10.1254/fpj.144.160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
|
48
|
Thoma G, Drückes P, Zerwes HG. Selective inhibitors of the Janus kinase Jak3—Are they effective? Bioorg Med Chem Lett 2014; 24:4617-4621. [DOI: 10.1016/j.bmcl.2014.08.046] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 08/19/2014] [Accepted: 08/20/2014] [Indexed: 01/21/2023]
|
49
|
Jasuja H, Chadha N, Kaur M, Silakari O. Pharmacophore and docking-based virtual screening approach for the design of new dual inhibitors of Janus kinase 1 and Janus kinase 2. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2014; 25:617-636. [PMID: 25148044 DOI: 10.1080/1062936x.2014.884163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Janus kinase 1 and 2, non-receptor protein tyrosine kinases, are implicated in various cancerous diseases. Involvement of these two enzymes in the pathways that stimulate cell proliferation in cancerous conditions makes them potential therapeutic targets for designing new dual-targeted agents for the treatment of cancer. In the present study, two separate pharmacophore models were developed and the best models for JAK1 (AAADH.25) and JAK2 (ADRR.92) were selected on the basis of their external predictive ability. Both models were subjected to a systematic virtual screening (VS) protocol using a PHASE database of 1.5 million molecules. The hits retrieved in VS were investigated for ADME properties to avoid selection of molecules with a poor pharmacokinetic profile. The molecules considered to be within the range of acceptable limits of ADME properties were further employed for docking simulations with JAK1 and JAK2 proteins to explore the final hits that possess structural features of both pharmacophore models as well as display essential interactions with both of them. Thus, the new molecules obtained in this way should show inhibitory activity against JAK1 and JAK2 and may serve as novel therapeutic agents for the treatment of cancerous disease conditions.
Collapse
Affiliation(s)
- H Jasuja
- a Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research , Punjabi University , Patiala , India
| | | | | | | |
Collapse
|
50
|
Gehringer M, Forster M, Pfaffenrot E, Bauer SM, Laufer SA. Novel hinge-binding motifs for Janus kinase 3 inhibitors: a comprehensive structure-activity relationship study on tofacitinib bioisosteres. ChemMedChem 2014; 9:2516-27. [PMID: 25139757 DOI: 10.1002/cmdc.201402252] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Indexed: 11/09/2022]
Abstract
The Janus kinases (JAKs) are a family of cytosolic tyrosine kinases crucially involved in cytokine signaling. JAKs have been demonstrated to be valid targets in the treatment of inflammatory and myeloproliferative disorders, and two inhibitors, tofacitinib and ruxolitinib, recently received their marketing authorization. Despite this success, selectivity within the JAK family remains a major issue. Both approved compounds share a common 7H-pyrrolo[2,3-d]pyrimidine hinge binding motif, and little is known about modifications tolerated at this heterocyclic core. In the current study, a library of tofacitinib bioisosteres was prepared and tested against JAK3. The compounds possessed the tofacitinib piperidinyl side chain, whereas the hinge binding motif was replaced by a variety of heterocycles mimicking its pharmacophore. In view of the promising expectations obtained from molecular modeling, most of the compounds proved to be poorly active. However, strategies for restoring activity within this series of novel chemotypes were discovered and crucial structure-activity relationships were deduced. The compounds presented may serve as starting point for developing novel JAK inhibitors and as a valuable training set for in silico models.
Collapse
Affiliation(s)
- Matthias Gehringer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical and Medicinal Chemistry, Eberhard Karls University of Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen (Germany), Fax: (+49) 7071-29-5037
| | | | | | | | | |
Collapse
|