1
|
Lv Y, Qi J, Babon JJ, Cao L, Fan G, Lang J, Zhang J, Mi P, Kobe B, Wang F. The JAK-STAT pathway: from structural biology to cytokine engineering. Signal Transduct Target Ther 2024; 9:221. [PMID: 39169031 PMCID: PMC11339341 DOI: 10.1038/s41392-024-01934-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/12/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024] Open
Abstract
The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway serves as a paradigm for signal transduction from the extracellular environment to the nucleus. It plays a pivotal role in physiological functions, such as hematopoiesis, immune balance, tissue homeostasis, and surveillance against tumors. Dysregulation of this pathway may lead to various disease conditions such as immune deficiencies, autoimmune diseases, hematologic disorders, and cancer. Due to its critical role in maintaining human health and involvement in disease, extensive studies have been conducted on this pathway, ranging from basic research to medical applications. Advances in the structural biology of this pathway have enabled us to gain insights into how the signaling cascade operates at the molecular level, laying the groundwork for therapeutic development targeting this pathway. Various strategies have been developed to restore its normal function, with promising therapeutic potential. Enhanced comprehension of these molecular mechanisms, combined with advances in protein engineering methodologies, has allowed us to engineer cytokines with tailored properties for targeted therapeutic applications, thereby enhancing their efficiency and safety. In this review, we outline the structural basis that governs key nodes in this pathway, offering a comprehensive overview of the signal transduction process. Furthermore, we explore recent advances in cytokine engineering for therapeutic development in this pathway.
Collapse
Affiliation(s)
- You Lv
- Center for Molecular Biosciences and Non-communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi, 710054, China
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi, 710026, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100080, China
| | - Jeffrey J Babon
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Longxing Cao
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Guohuang Fan
- Immunophage Biotech Co., Ltd, No. 10 Lv Zhou Huan Road, Shanghai, 201112, China
| | - Jiajia Lang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jin Zhang
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi, 710026, China
| | - Pengbing Mi
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Queensland, 4072, Australia.
| | - Faming Wang
- Center for Molecular Biosciences and Non-communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi, 710054, China.
| |
Collapse
|
2
|
Kiełbowski K, Plewa P, Bratborska AW, Bakinowska E, Pawlik A. JAK Inhibitors in Rheumatoid Arthritis: Immunomodulatory Properties and Clinical Efficacy. Int J Mol Sci 2024; 25:8327. [PMID: 39125897 PMCID: PMC11311960 DOI: 10.3390/ijms25158327] [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: 06/19/2024] [Revised: 07/20/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
Rheumatoid arthritis (RA) is a highly prevalent autoimmune disorder. The pathogenesis of the disease is complex and involves various cellular populations, including fibroblast-like synoviocytes, macrophages, and T cells, among others. Identification of signalling pathways and molecules that actively contribute to the development of the disease is crucial to understanding the mechanisms involved in the chronic inflammatory environment present in affected joints. Recent studies have demonstrated that the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway regulates the behaviour of immune cells and contributes to the progression of RA. Several JAK inhibitors, such as tofacitinib, baricitinib, upadacitinib, and filgocitinib, have been developed, and their efficacy and safety in patients with RA have been comprehensively investigated in a number of clinical trials. Consequently, JAK inhibitors have been approved and registered as a treatment for patients with RA. In this review, we discuss the involvement of JAK/STAT signalling in the pathogenesis of RA and summarise the potential beneficial effects of JAK inhibitors in cells implicated in the pathogenesis of the disease. Moreover, we present the most important phase 3 clinical trials that evaluated the use of these agents in patients.
Collapse
Affiliation(s)
- Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.)
| | - Paulina Plewa
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland;
| | | | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.)
| |
Collapse
|
3
|
Khokhar M, Dey S, Tomo S, Jaremko M, Emwas AH, Pandey RK. Unveiling Novel Drug Targets and Emerging Therapies for Rheumatoid Arthritis: A Comprehensive Review. ACS Pharmacol Transl Sci 2024; 7:1664-1693. [PMID: 38898941 PMCID: PMC11184612 DOI: 10.1021/acsptsci.4c00067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic debilitating autoimmune disease, that causes joint damage, deformities, and decreased functionality. In addition, RA can also impact organs like the skin, lungs, eyes, and blood vessels. This autoimmune condition arises when the immune system erroneously targets the joint synovial membrane, resulting in synovitis, pannus formation, and cartilage damage. RA treatment is often holistic, integrating medication, physical therapy, and lifestyle modifications. Its main objective is to achieve remission or low disease activity by utilizing a "treat-to-target" approach that optimizes drug usage and dose adjustments based on clinical response and disease activity markers. The primary RA treatment uses disease-modifying antirheumatic drugs (DMARDs) that help to interrupt the inflammatory process. When there is an inadequate response, a combination of biologicals and DMARDs is recommended. Biological therapies target inflammatory pathways and have shown promising results in managing RA symptoms. Close monitoring for adverse effects and disease progression is critical to ensure optimal treatment outcomes. A deeper understanding of the pathways and mechanisms will allow new treatment strategies that minimize adverse effects and maintain quality of life. This review discusses the potential targets that can be used for designing and implementing precision medicine in RA treatment, spotlighting the latest breakthroughs in biologics, JAK inhibitors, IL-6 receptor antagonists, TNF blockers, and disease-modifying noncoding RNAs.
Collapse
Affiliation(s)
- Manoj Khokhar
- Department
of Biochemistry, All India Institute of
Medical Sciences, Jodhpur, 342005 Rajasthan, India
| | - Sangita Dey
- CSO
Department, Cellworks Research India Pvt
Ltd, Bengaluru, 560066 Karnataka, India
| | - Sojit Tomo
- Department
of Biochemistry, All India Institute of
Medical Sciences, Jodhpur, 342005 Rajasthan, India
| | - Mariusz Jaremko
- Smart-Health
Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological
and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955 Jeddah, Saudi Arabia
| | - Abdul-Hamid Emwas
- Core
Laboratories, King Abdullah University of
Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Rajan Kumar Pandey
- Department
of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm 17177, Sweden
| |
Collapse
|
4
|
Chen L, Tang Y, Lang JJ, Lin Y, Yu Z, Li X, Zheng X, Mi P, Lv Y, Lin YW. Design, synthesis and evaluation of C-5 substituted pyrrolopyridine derivatives as potent Janus Kinase 1 inhibitors with excellent selectivity. Eur J Med Chem 2024; 267:116210. [PMID: 38359535 DOI: 10.1016/j.ejmech.2024.116210] [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: 12/17/2023] [Revised: 01/15/2024] [Accepted: 01/31/2024] [Indexed: 02/17/2024]
Abstract
The development of highly selective Janus Kinase 1 (JAK1) inhibitors is crucial for improving efficacy and minimizing adverse effects in the clinical treatment of autoimmune diseases. In a prior study, we designed a series of C-5 4-pyrazol substituted pyrrolopyridine derivatives that demonstrated significant potency against JAK1, with a 10 ∼ 20-fold selectivity over Janus Kinase 2 (JAK2). Building on this foundation, we adopted orthogonal strategy by modifying the C-5 position with 3-pyrazol/4-pyrazol/3-pyrrol groups and tail with substituted benzyl groups on the pyrrolopyridine head to enhance both potency and selectivity. In this endeavor, we have identified several compounds that exhibit excellent potency and selectivity for JAK1. Notably, compounds 12b and 12e, which combined 4-pyrazol group at C-5 site and meta-substituted benzyl tails, displayed IC50 value with 2.4/2.2 nM and high 352-/253-fold selectivity for JAK1 over JAK2 in enzyme assays. Additionally, both compounds showed good JAK1-selective in Ba/F3-TEL-JAK1/2 cell-based assays. These findings mark a substantial improvement, as these compounds are 10-fold more potent and over 10-fold more selective than the best compound identified in our previous study. The noteworthy potency and selectivity properties of compounds 12b and 12e suggest their potential utility in furthering the development of drugs for autoimmune diseases.
Collapse
Affiliation(s)
- Limei Chen
- Department of Pharmacy, Hengyang Medicinal School, University of South China, Hengyang, Hunan, 421001, China
| | - Yahua Tang
- The Affiliated Nanhua Hospital, Department of Pharmacy, Institute of Clinical Pharmacy, Hengyang Medical School, University of South China, Hunan, 421001, China
| | - Jia-Jia Lang
- School of Chemistry and Chemical Engineering, University of South China, Hengyang, Hunan, 421001, China; Key Lab of Protein Structure and Function of Universities in Hunan Province, University of South China, Hengyang, Hunan, 421001, China
| | - Yuqing Lin
- Department of Pharmacy, Hengyang Medicinal School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhixin Yu
- Department of Pharmacy, Hengyang Medicinal School, University of South China, Hengyang, Hunan, 421001, China
| | - Xinhao Li
- Department of Pharmacy, Hengyang Medicinal School, University of South China, Hengyang, Hunan, 421001, China
| | - Xing Zheng
- Department of Pharmacy, Hengyang Medicinal School, University of South China, Hengyang, Hunan, 421001, China; Department of Pharmacy, Hunan Vocational College of Science and Technology, Changsha, Hunan, 410004, China
| | - Pengbing Mi
- Department of Pharmacy, Hengyang Medicinal School, University of South China, Hengyang, Hunan, 421001, China; Key Lab of Protein Structure and Function of Universities in Hunan Province, University of South China, Hengyang, Hunan, 421001, China.
| | - You Lv
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, Shaanxi, 710021, China; Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi, 710026, China.
| | - Ying-Wu Lin
- School of Chemistry and Chemical Engineering, University of South China, Hengyang, Hunan, 421001, China; Key Lab of Protein Structure and Function of Universities in Hunan Province, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
5
|
Yuan Y, Zhang XF, Li YC, Chen HQ, Wen T, Zheng JL, Zhao ZY, Hu QY. VX-509 attenuates the stemness characteristics of colorectal cancer stem-like cells by regulating the epithelial-mesenchymal transition through Nodal/Smad2/3 signaling. World J Stem Cells 2024; 16:207-227. [PMID: 38455101 PMCID: PMC10915959 DOI: 10.4252/wjsc.v16.i2.207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/19/2023] [Accepted: 01/16/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Colorectal cancer stem cells (CCSCs) are heterogeneous cells that can self-renew and undergo multidirectional differentiation in colorectal cancer (CRC) patients. CCSCs are generally accepted to be important sources of CRC and are responsible for the progression, metastasis, and therapeutic resistance of CRC. Therefore, targeting this specific subpopulation has been recognized as a promising strategy for overcoming CRC. AIM To investigate the effect of VX-509 on CCSCs and elucidate the underlying mechanism. METHODS CCSCs were enriched from CRC cell lines by in conditioned serum-free medium. Western blot, Aldefluor, transwell and tumorigenesis assays were performed to verify the phenotypic characteristics of the CCSCs. The anticancer efficacy of VX-509 was assessed in HCT116 CCSCs and HT29 CCSCs by performing cell viability analysis, colony formation, sphere formation, flow cytometry, and western blotting assessments in vitro and tumor growth, immunohistochemistry and immunofluorescence assessments in vivo. RESULTS Compared with parental cells, sphere cells derived from HCT116 and HT29 cells presented increased expression of stem cell transcription factors and stem cell markers and were more potent at promoting migration and tumorigenesis, demonstrating that the CRC sphere cells displayed CSC features. VX-509 inhibited the tumor malignant biological behavior of CRC-stem-like cells, as indicated by their proliferation, migration and clonality in vitro, and suppressed the tumor of CCSC-derived xenograft tumors in vivo. Besides, VX-509 suppressed the CSC characteristics of CRC-stem-like cells and inhibited the progression of epithelial-mesenchymal transition (EMT) signaling in vitro. Nodal was identified as the regulatory factor of VX-509 on CRC stem-like cells through analyses of differentially expressed genes and CSC-related database information. VX-509 markedly downregulated the expression of Nodal and its downstream phosphorylated Smad2/3 to inhibit EMT progression. Moreover, VX-509 reversed the dedifferentiation of CCSCs and inhibited the progression of EMT induced by Nodal overexpression. CONCLUSION VX-509 prevents the EMT process in CCSCs by inhibiting the transcription and protein expression of Nodal, and inhibits the dedifferentiated self-renewal of CCSCs.
Collapse
Affiliation(s)
- Yun Yuan
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| | - Xu-Fan Zhang
- Department of Nuclear Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China
| | - Yu-Chen Li
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| | - Hong-Qing Chen
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| | - Tian Wen
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| | - Jia-Lian Zheng
- Department of Hepatology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110032, Liaoning Province, China
| | - Zi-Yi Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
- Traditional Chinese Medicine Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, Sichuan Province, China
| | - Qiong-Ying Hu
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China.
| |
Collapse
|
6
|
Pang Q, You L, Meng X, Li Y, Deng T, Li D, Zhu B. Regulation of the JAK/STAT signaling pathway: The promising targets for cardiovascular disease. Biochem Pharmacol 2023; 213:115587. [PMID: 37187275 DOI: 10.1016/j.bcp.2023.115587] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
Individuals have known that Janus kinase (JAK) signal transducer and activator of transcription (STAT) signaling pathway was involved in the growth of the cell, cell differentiation courses advancement, immune cellular survival, as well as hematopoietic system advancement. Researches in the animal models have already uncovered a JAK/STAT regulatory function in myocardial ischemia-reperfusion injury (MIRI), acute myocardial infarction (MI), hypertension, myocarditis, heart failure, angiogenesis and fibrosis. Evidences originating in these studies indicate a therapeutic JAK/STAT function in cardiovascular diseases (CVDs). In this retrospection, various JAK/STAT functions in the normal and ill hearts were described. Moreover, the latest figures about JAK/STAT were summarized under the background of CVDs. Finally, we discussed the clinical transformation prospects and technical limitations of JAK/STAT as the potential therapeutic targets for CVDs. This collection of evidences has essential meanings for the clinical application of JAK/STAT as medicinal agents for CVDs. In this retrospection, various JAK/STAT functions in the normal and ill hearts were described. Moreover, the latest figures about JAK/STAT were summarized under the background of CVDs. Finally, we discussed the clinical transformation prospects and toxicity of JAK/STAT inhibitors as potential therapeutic targets for CVDs. This collection of evidences has essential meanings for the clinical application of JAK/STAT as medicinal agents for CVDs.
Collapse
Affiliation(s)
- Qiuyu Pang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lu You
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangmin Meng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yumeng Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Deng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Deyong Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Bingmei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
7
|
Xue C, Yao Q, Gu X, Shi Q, Yuan X, Chu Q, Bao Z, Lu J, Li L. Evolving cognition of the JAK-STAT signaling pathway: autoimmune disorders and cancer. Signal Transduct Target Ther 2023; 8:204. [PMID: 37208335 DOI: 10.1038/s41392-023-01468-7] [Citation(s) in RCA: 166] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/22/2023] [Indexed: 05/21/2023] Open
Abstract
The Janus kinase (JAK) signal transducer and activator of transcription (JAK-STAT) pathway is an evolutionarily conserved mechanism of transmembrane signal transduction that enables cells to communicate with the exterior environment. Various cytokines, interferons, growth factors, and other specific molecules activate JAK-STAT signaling to drive a series of physiological and pathological processes, including proliferation, metabolism, immune response, inflammation, and malignancy. Dysregulated JAK-STAT signaling and related genetic mutations are strongly associated with immune activation and cancer progression. Insights into the structures and functions of the JAK-STAT pathway have led to the development and approval of diverse drugs for the clinical treatment of diseases. Currently, drugs have been developed to mainly target the JAK-STAT pathway and are commonly divided into three subtypes: cytokine or receptor antibodies, JAK inhibitors, and STAT inhibitors. And novel agents also continue to be developed and tested in preclinical and clinical studies. The effectiveness and safety of each kind of drug also warrant further scientific trials before put into being clinical applications. Here, we review the current understanding of the fundamental composition and function of the JAK-STAT signaling pathway. We also discuss advancements in the understanding of JAK-STAT-related pathogenic mechanisms; targeted JAK-STAT therapies for various diseases, especially immune disorders, and cancers; newly developed JAK inhibitors; and current challenges and directions in the field.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qinfan Yao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinyu Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhengyi Bao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
8
|
Lang JJ, Lv Y, Kobe B, Chen H, Tan Y, Chen L, Wang X, Mi P, Zheng X, Lin YW. Discovery of C-5 Pyrazole-Substituted Pyrrolopyridine Derivatives as Potent and Selective Inhibitors for Janus Kinase 1. J Med Chem 2023; 66:6725-6742. [PMID: 37163463 DOI: 10.1021/acs.jmedchem.3c00050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Developing selective inhibitors for Janus kinase 1 (JAK1) is a significant focus for improving the efficacy and alleviating the adverse effects in treating immune-inflammatory diseases. Herein, we report the discovery of a series of C-5 pyrazole-modified pyrrolopyrimidine derivatives as JAK1-selective inhibitors. The potential hydrogen bond between the pyrazole group and E966 in JAK1 is the key point that enhances JAK1 selectivity. These compounds exhibit 10- to 20-fold JAK1 selectivity over JAK2 in enzyme assays. Compound 12b also exhibits excellent JAK1 selectivity in Ba/F3-TEL-JAK cellular assays. Metabolism studies and the results of the hair growth model in mice indicate that compound 12b may be a viable lead compound for the development of highly JAK1-selective inhibitors for immune and inflammatory diseases.
Collapse
Affiliation(s)
- Jia-Jia Lang
- Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hengyang Medical College, School of Chemistry and Chemical Engineering, University of South China, Hengyang 421001, China
- Key Lab of Protein Structure and Function of Universities in Hunan Province, University of South China, Hengyang 421001, China
| | - You Lv
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, Shaanxi 710021, China
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi 710026, China
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Hongfei Chen
- Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yan Tan
- Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Limei Chen
- Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xuechuan Wang
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, Shaanxi 710021, China
| | - Pengbing Mi
- Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Key Lab of Protein Structure and Function of Universities in Hunan Province, University of South China, Hengyang 421001, China
| | - Xing Zheng
- Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Pharmacy, Hunan Vocational College of Science and Technology, Changsha 410004, China
| | - Ying-Wu Lin
- Hengyang Medical College, School of Chemistry and Chemical Engineering, University of South China, Hengyang 421001, China
- Key Lab of Protein Structure and Function of Universities in Hunan Province, University of South China, Hengyang 421001, China
| |
Collapse
|
9
|
Chen X, Zhang L, Bao Q, Meng F, Liu C, Xu R, Ji X, You Q, Jiang Z. A JAK tyrosine kinase and pseudokinase Co-inhibition strategy combines enhanced potency and on-demand activation. Eur J Med Chem 2023; 250:115198. [PMID: 36805946 DOI: 10.1016/j.ejmech.2023.115198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/02/2023] [Accepted: 02/09/2023] [Indexed: 02/12/2023]
Abstract
Janus tyrosine kinase (JAK) inhibitors have been on the market for several years, but their use is limited by drug resistance and intolerable side effects. Herein, we propose a novel strategy of JAK tyrosine kinase (TK) and pseudokinase (PK) domain co-inhibition system to consolidate robust JAK inhibition and on-demand activation. A photoexcited prodrug PAT-SIL-TG-1&AT exhibits the synergy effects of TK-PK co-inhibition and enable the spatiotemporal control of JAK2 signaling. The hypoxia-activated prodrug HAT-SIL-TG-1&AT significantly inhibited HEL cells proliferation and downregulated phosphorylated STAT3/5 under hypoxic conditions. Importantly, HAT-SIL-TG-1&AT showed synergistic antitumor effects and selectively inhibited the JAK-STAT signaling in tumor tissues in vivo. This work demonstrates a viable solution to achieve superior JAK2 inhibition, and provides an inspiration for other kinases containing PK domain.
Collapse
Affiliation(s)
- Xuetao Chen
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Liangying Zhang
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Pharmacy, Hunan Food and Drug Vocational College, Changsha, 410208, China
| | - Qichao Bao
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Fanying Meng
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Chihong Liu
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Rujun Xu
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xinrui Ji
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qidong You
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Zhengyu Jiang
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
10
|
Signaling pathways in rheumatoid arthritis: implications for targeted therapy. Signal Transduct Target Ther 2023; 8:68. [PMID: 36797236 PMCID: PMC9935929 DOI: 10.1038/s41392-023-01331-9] [Citation(s) in RCA: 152] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/16/2022] [Accepted: 01/18/2023] [Indexed: 02/18/2023] Open
Abstract
Rheumatoid arthritis (RA) is an incurable systemic autoimmune disease. Disease progression leads to joint deformity and associated loss of function, which significantly impacts the quality of life for sufferers and adds to losses in the labor force. In the past few decades, RA has attracted increased attention from researchers, the abnormal signaling pathways in RA are a very important research field in the diagnosis and treatment of RA, which provides important evidence for understanding this complex disease and developing novel RA-linked intervention targets. The current review intends to provide a comprehensive overview of RA, including a general introduction to the disease, historical events, epidemiology, risk factors, and pathological process, highlight the primary research progress of the disease and various signaling pathways and molecular mechanisms, including genetic factors, epigenetic factors, summarize the most recent developments in identifying novel signaling pathways in RA and new inhibitors for treating RA. therapeutic interventions including approved drugs, clinical drugs, pre-clinical drugs, and cutting-edge therapeutic technologies. These developments will hopefully drive progress in new strategically targeted therapies and hope to provide novel ideas for RA treatment options in the future.
Collapse
|
11
|
Wang Q, Chen D, Wang Y, Dong C, Liu J, Chen K, Song F, Wang C, Yuan J, Davis RA, Kuek V, Jin H, Xu J. Thiaplakortone B attenuates RANKL-induced NF-κB and MAPK signaling and dampens OVX-induced bone loss in mice. Biomed Pharmacother 2022; 154:113622. [PMID: 36081291 DOI: 10.1016/j.biopha.2022.113622] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/24/2022] [Accepted: 08/29/2022] [Indexed: 12/16/2022] Open
Abstract
Osteoclasts play an important role in maintaining the relative stability of bone mass. Abnormal number and function of osteoclasts are closely related to osteoporosis and osteolytic diseases. Thiaplakortone B (TPB), a natural compound derived from the Great Barrier Reef sponge Plakortis lita, has been reported to inhibit the growth of the malaria parasite, Plasmodium falciparum, but its effect on osteoclastogenesis has not been previously investigated. In our study, we found that TPB suppresses the receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-induced osteoclast formation and resorption activity by tartrate-resistant acid phosphatase (TRAcP) staining, immunofluorescence staining of F-actin belts and hydroxyapatite resorption assay. Furthermore, using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting analysis, we discovered that TPB inhibits osteoclast-specific genes and proteins expression. Mechanistically, TPB blocks multiple upstream pathways including calcium oscillation, NF-κB, mitogen-activated protein kinase (MAPK) and nuclear factor of activated T cells 1(NFATc1) signaling pathways. In vivo, TPB could dampen bone loss in an ovariectomy (OVX) mouse model by micro-CT assessment and histological staining. Therefore, TPB may serve as a potential therapeutic candidate for the treatment of osteoporosis and osteolysis.
Collapse
Affiliation(s)
- Qingqing Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang 310016, China; School of Biomedical Sciences, University of Western Australia, Perth, Western Australia 6009, Australia
| | - Delong Chen
- School of Biomedical Sciences, University of Western Australia, Perth, Western Australia 6009, Australia
| | - Yining Wang
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chenlin Dong
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jian Liu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Kai Chen
- School of Biomedical Sciences, University of Western Australia, Perth, Western Australia 6009, Australia
| | - Fangming Song
- Research Centre for Regenerative Medicine and Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Guangxi, 530021, China
| | - Chao Wang
- School of Biomedical Sciences, University of Western Australia, Perth, Western Australia 6009, Australia
| | - Jinbo Yuan
- School of Biomedical Sciences, University of Western Australia, Perth, Western Australia 6009, Australia
| | - Rohan A Davis
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Queensland, 4111, Australia
| | - Vincent Kuek
- School of Biomedical Sciences, University of Western Australia, Perth, Western Australia 6009, Australia.
| | - Haiming Jin
- School of Biomedical Sciences, University of Western Australia, Perth, Western Australia 6009, Australia; The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Jiake Xu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang 310016, China; School of Biomedical Sciences, University of Western Australia, Perth, Western Australia 6009, Australia.
| |
Collapse
|
12
|
Laux J, Forster M, Riexinger L, Schwamborn A, Guezguez J, Pokoj C, Kudolo M, Berger LM, Knapp S, Schollmeyer D, Guse J, Burnet M, Laufer SA. Pharmacokinetic Optimization of Small Molecule Janus Kinase 3 Inhibitors to Target Immune Cells. ACS PHARMACOLOGY & TRANSLATIONAL SCIENCE 2022; 5:573-602. [DOI: 10.1021/acsptsci.2c00054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Julian Laux
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Michael Forster
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE Germany
| | - Laura Riexinger
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Anna Schwamborn
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Jamil Guezguez
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Christina Pokoj
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Mark Kudolo
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE Germany
| | - Lena M. Berger
- Structural Genomics Consortium, Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Stefan Knapp
- Structural Genomics Consortium, Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Dieter Schollmeyer
- Institute for Organic Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55099 Mainz, Germany
| | - Jan Guse
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Michael Burnet
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Stefan A. Laufer
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
- Tübingen Center for Academic Drug Discovery and Development (TüCAD2), 72076 Tübingen, Germany
| |
Collapse
|
13
|
Srivastava S, Rasool M. Underpinning IL-6 biology and emphasizing selective JAK blockade as the potential alternate therapeutic intervention for rheumatoid arthritis. Life Sci 2022; 298:120516. [DOI: 10.1016/j.lfs.2022.120516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/08/2022] [Accepted: 03/25/2022] [Indexed: 02/07/2023]
|
14
|
Xu Z, Chu M. Advances in Immunosuppressive Agents Based on Signal Pathway. Front Pharmacol 2022; 13:917162. [PMID: 35694243 PMCID: PMC9178660 DOI: 10.3389/fphar.2022.917162] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/02/2022] [Indexed: 12/13/2022] Open
Abstract
Immune abnormality involves in various diseases, such as infection, allergic diseases, autoimmune diseases, as well as transplantation. Several signal pathways have been demonstrated to play a central role in the immune response, including JAK/STAT, NF-κB, PI3K/AKT-mTOR, MAPK, and Keap1/Nrf2/ARE pathway, in which multiple targets have been used to develop immunosuppressive agents. In recent years, varieties of immunosuppressive agents have been approved for clinical use, such as the JAK inhibitor tofacitinib and the mTOR inhibitor everolimus, which have shown good therapeutic effects. Additionally, many immunosuppressive agents are still in clinical trials or preclinical studies. In this review, we classified the immunosuppressive agents according to the immunopharmacological mechanisms, and summarized the phase of immunosuppressive agents.
Collapse
Affiliation(s)
- Zhiqing Xu
- Department of Immunology, National Health Commission (NHC) Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Pharmacology, Jilin University, Changchun, China
| | - Ming Chu
- Department of Immunology, National Health Commission (NHC) Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
15
|
Zhao MY, Zhang W, Rao GW. Targeting Janus Kinase (JAK) for Fighting Diseases: The Research of JAK Inhibitor Drugs. Curr Med Chem 2022; 29:5010-5040. [PMID: 35255783 DOI: 10.2174/1568026622666220307124142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/11/2021] [Accepted: 12/21/2021] [Indexed: 11/22/2022]
Abstract
Janus Kinase (JAK), a nonreceptor protein tyrosine kinase, has emerged as an excellent target through research and development since its discovery in the 1990s. As novel small-molecule targeted drugs, JAK inhibitor drugs have been successfully used in the treatment of rheumatoid arthritis (RA), myofibrosis (MF) and ulcerative colitis (UC). With the gradual development of JAK targets in the market, JAK inhibitors have also received very considerable feedback in the treatment of autoimmune diseases such as atopic dermatitis (AD), Crohn's disease (CD) and graft-versus host disease (GVHD). This article reviews the research progress of JAK inhibitor drugs: introducing the existing JAK inhibitors on the market and some JAK inhibitors in clinical trials currently. In addition, the synthesis of various types of JAK inhibitors were summarized, and the effects of different drug structures on drug inhibition and selectivity.
Collapse
Affiliation(s)
- Min-Yan Zhao
- College of Pharmaceutical Science, Zhejiang University of Technology, and Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Wen Zhang
- College of Pharmaceutical Science, Zhejiang University of Technology, and Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Guo-Wu Rao
- College of Pharmaceutical Science, Zhejiang University of Technology, and Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| |
Collapse
|
16
|
Alharthi S, Turkmani M, AlJasser MI. Acne exacerbation after tofacitinib treatment for alopecia areata. Dermatol Reports 2022; 14:9396. [PMID: 35832266 PMCID: PMC9272012 DOI: 10.4081/dr.2022.9396] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 10/12/2021] [Indexed: 11/23/2022] Open
Abstract
The major discovery of the novel therapeutic Janus kinase (JAK) inhibitors has been implicated in several dermatological diseases. Recently, studies have shown the efficacy and encouraging results of oral JAK inhibitors as a treatment for alopecia areata (AA). Due to the novelty of this treatment, potential side effects are not fully explored. In this paper, we present a case of a 28-year-old male with a 10-year history of alopecia totalis (AT) treated successfully with tofacitinib with encouraging effects on hair regrowth; however, a significant worsening of the patient’s facial acne was observed four months after AT treatment initiation. JAK inhibitors have promising results in the management of different dermatological conditions including moderatesevere forms of AA with few reported adverse events. Acne exacerbation is a unique observed adverse effect of this therapy. More thorough larger sized studies are needed to further characterize the association between acne exacerbation and the use of JAK inhibitors.
Collapse
|
17
|
Hu X, Li J, Fu M, Zhao X, Wang W. The JAK/STAT signaling pathway: from bench to clinic. Signal Transduct Target Ther 2021; 6:402. [PMID: 34824210 PMCID: PMC8617206 DOI: 10.1038/s41392-021-00791-1] [Citation(s) in RCA: 1168] [Impact Index Per Article: 292.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/09/2021] [Accepted: 09/21/2021] [Indexed: 02/08/2023] Open
Abstract
The Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway was discovered more than a quarter-century ago. As a fulcrum of many vital cellular processes, the JAK/STAT pathway constitutes a rapid membrane-to-nucleus signaling module and induces the expression of various critical mediators of cancer and inflammation. Growing evidence suggests that dysregulation of the JAK/STAT pathway is associated with various cancers and autoimmune diseases. In this review, we discuss the current knowledge about the composition, activation, and regulation of the JAK/STAT pathway. Moreover, we highlight the role of the JAK/STAT pathway and its inhibitors in various diseases.
Collapse
Affiliation(s)
- Xiaoyi Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu, 610041, Sichuan, P. R. China
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, 610041, Chengdu, P. R. China
| | - Jing Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu, 610041, Sichuan, P. R. China
| | - Maorong Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu, 610041, Sichuan, P. R. China
| | - Xia Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu, 610041, Sichuan, P. R. China.
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, 610041, Chengdu, P. R. China.
| | - Wei Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu, 610041, Sichuan, P. R. China.
| |
Collapse
|
18
|
Second-Generation Jak2 Inhibitors for Advanced Prostate Cancer: Are We Ready for Clinical Development? Cancers (Basel) 2021; 13:cancers13205204. [PMID: 34680353 PMCID: PMC8533841 DOI: 10.3390/cancers13205204] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/05/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Prostate Cancer (PC) is currently estimated to affect 1 in 9 men and is the second leading cause of cancer in men in the US. While androgen deprivation therapy, which targets the androgen receptor, is one of the front-line therapies for advanced PC and for recurrence of organ-confined PC treated with surgery, lethal castrate-resistant PC develops consistently in patients. PC is a multi-focal cancer with different grade carcinoma areas presenting simultaneously. Jak2-Stat5 signaling pathway has emerged as a potentially highly effective molecular target in PCs with positive areas for activated Stat5 protein. Activated Jak2-Stat5 signaling can be readily targeted by the second-generation Jak2-inhibitors that have been developed for myeloproliferative and autoimmune disorders and hematological malignancies. In this review, we analyze and summarize the Jak2 inhibitors that are currently in preclinical and clinical development. Abstract Androgen deprivation therapy (ADT) for metastatic and high-risk prostate cancer (PC) inhibits growth pathways driven by the androgen receptor (AR). Over time, ADT leads to the emergence of lethal castrate-resistant PC (CRPC), which is consistently caused by an acquired ability of tumors to re-activate AR. This has led to the development of second-generation anti-androgens that more effectively antagonize AR, such as enzalutamide (ENZ). However, the resistance of CRPC to ENZ develops rapidly. Studies utilizing preclinical models of PC have established that inhibition of the Jak2-Stat5 signaling leads to extensive PC cell apoptosis and decreased tumor growth. In large clinical cohorts, Jak2-Stat5 activity predicts PC progression and recurrence. Recently, Jak2-Stat5 signaling was demonstrated to induce ENZ-resistant PC growth in preclinical PC models, further emphasizing the importance of Jak2-Stat5 for therapeutic targeting for advanced PC. The discovery of the Jak2V617F somatic mutation in myeloproliferative disorders triggered the rapid development of Jak1/2-specific inhibitors for a variety of myeloproliferative and auto-immune disorders as well as hematological malignancies. Here, we review Jak2 inhibitors targeting the mutated Jak2V617F vs. wild type (WT)-Jak2 that are currently in the development pipeline. Among these 35 compounds with documented Jak2 inhibitory activity, those with potency against WT-Jak2 hold strong potential for advanced PC therapy.
Collapse
|
19
|
Sonkar C, Doharey PK, Rathore AS, Singh V, Kashyap D, Sahoo AK, Mittal N, Sharma B, Jha HC. Repurposing of gastric cancer drugs against COVID-19. Comput Biol Med 2021; 137:104826. [PMID: 34537409 PMCID: PMC8420180 DOI: 10.1016/j.compbiomed.2021.104826] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 12/19/2022]
Abstract
Corona Virus Disease 2019 (COVID-19) caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has become a global pandemic. Additionally, the SARS-CoV-2 infection in the patients of Gastric Cancer (GC; the third leading cause of death in the world) pose a great challenge for the health management of the patients. Since there have been uncertainties to develop a new drug against COVID-19, there is an urgent need for repurposing drugs that can target key proteins of both SARS-CoV-2 and GC. The SARS-CoV-2-RdRp protein contains the NiRAN domain, which is known to have kinase-like folds. A docking study of the FDA approved drugs against GC was performed using AutoDock 4.2 and Glide Schrodinger suite 2019 against SARS-CoV-2-RdRp protein. MMGBSA and MD simulation studies were performed to investigate the binding and stability of the inhibitors with the target protein. In this study, we have found 12 kinase inhibitors with high binding energies namely Baricitinib, Brepocitinib, Decernotinib, Fasudil, Filgotinib, GSK2606414, Peficitinib, Ruxolitinib, Tofacitinib, Upadacitinib, Pamapimod and Ibrutinib. These FDA approved drugs against GC can play a key role in the treatment of COVID-19 patients along with GC as comorbidity. We also hypothesize that JAK, ITK, Rho-associated kinases, FGFR2, FYN, PERK, TYK2, p38-MAPK and SYK kinases can be considered as key therapeutic targets in COVID-19 treatment. Taken altogether, we have proposed the SARS-CoV-2-RdRp as a potential therapeutic target through in-silico studies. However, further in-vitro and in-vivo studies are required for the validation of the proposed targets and drugs for the treatment of COVID-19 patients already suffering from GC.
Collapse
Affiliation(s)
- Charu Sonkar
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore, 453552, India
| | - Pawan Kumar Doharey
- Department of Biochemistry, University of Allahabad, Allahabad, 211002, U.P., India
| | - Anuranjan Singh Rathore
- SASTRA Deemed to Be University, Trichy-Tanjore Road, Thirumalaisamudram, Thanjavur, Tamil Nadu, 613401, India
| | - Vishal Singh
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Allahabad, 211015, U.P., India
| | - Dharmendra Kashyap
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore, 453552, India
| | - Amaresh Kumar Sahoo
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Allahabad, 211015, U.P., India
| | - Nitish Mittal
- Computational and Systems Biology, Biozentrum, University of Basel, Klingelbergstrasse 50-70, 4056, Basel, Switzerland
| | - Bechan Sharma
- Department of Biochemistry, University of Allahabad, Allahabad, 211002, U.P., India
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore, 453552, India.
| |
Collapse
|
20
|
Zhang L, Zhang Y, Pan J. Immunopathogenic mechanisms of rheumatoid arthritis and the use of anti-inflammatory drugs. Intractable Rare Dis Res 2021; 10:154-164. [PMID: 34466337 PMCID: PMC8397820 DOI: 10.5582/irdr.2021.01022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/25/2021] [Accepted: 04/02/2021] [Indexed: 11/05/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, progressive autoimmune disease characterized by synovitis and symmetrical joint destruction. RA has become one of the key diseases endangering human health, but its etiology is not clear. Therefore, identifying the immunopathogenic mechanisms of RA and developing therapeutic drugs to treat autoimmune diseases have always been difficult. This article mainly reviews the immunopathogenic mechanism of RA and advances in the study of anti-inflammatory drugs in order to provide a reference for the treatment of RA and drug development in the future.
Collapse
Affiliation(s)
- Ling Zhang
- Biomedical Sciences College, Shandong Medicinal Biotechnology Centre, Shandong First Medical University, Ji'nan, China
- Key Lab for Biotech-Drugs of National Health Commission, Shandong First Medical University, Ji'nan, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University, Ji'nan, China
| | - Yihang Zhang
- Biomedical Sciences College, Shandong Medicinal Biotechnology Centre, Shandong First Medical University, Ji'nan, China
- Key Lab for Biotech-Drugs of National Health Commission, Shandong First Medical University, Ji'nan, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University, Ji'nan, China
| | - Jihong Pan
- Biomedical Sciences College, Shandong Medicinal Biotechnology Centre, Shandong First Medical University, Ji'nan, China
- Key Lab for Biotech-Drugs of National Health Commission, Shandong First Medical University, Ji'nan, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University, Ji'nan, China
- Address correspondence to:Pan Jihong, Biomedical Sciences College, Shandong Medicinal Biotechnology Centre, Shandong First Medical University, # 6699 Qingdao Road, Ji'nan 250117, China. E-mail:
| |
Collapse
|
21
|
Dai Z, Chen J, Chang Y, Christiano AM. Selective inhibition of JAK3 signaling is sufficient to reverse alopecia areata. JCI Insight 2021; 6:142205. [PMID: 33830087 PMCID: PMC8119218 DOI: 10.1172/jci.insight.142205] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 02/25/2021] [Indexed: 12/19/2022] Open
Abstract
The Janus kinase/signal transducers and activators of transcription (JAK/STAT) are key intracellular mediators in the signal transduction of many cytokines and growth factors. Common γ chain cytokines and interferon-γ that use the JAK/STAT pathway to induce biological responses have been implicated in the pathogenesis of alopecia areata (AA), a T cell-mediated autoimmune disease of the hair follicle. We previously showed that therapeutic targeting of JAK/STAT pathways using the first-generation JAK1/2 inhibitor, ruxolitinib, and the pan-JAK inhibitor, tofacitinib, was highly effective in the treatment of human AA, as well as prevention and reversal of AA in the C3H/HeJ mouse model. To better define the role of individual JAKs in the pathogenesis of AA, in this study, we tested and compared the efficacy of several next-generation JAK-selective inhibitors in the C3H/HeJ mouse model of AA, using both systemic and topical delivery. We found that JAK1-selective inhibitors as well as JAK3-selective inhibitors robustly induced hair regrowth and decreased AA-associated inflammation, whereas several JAK2-selective inhibitors failed to restore hair growth in treated C3H/HeJ mice with AA. Unlike JAK1, which is broadly expressed in many tissues, JAK3 expression is largely restricted to hematopoietic cells. Our study demonstrates inhibiting JAK3 signaling is sufficient to prevent and reverse disease in the preclinical model of AA.
Collapse
Affiliation(s)
| | | | | | - Angela M. Christiano
- Department of Dermatology and
- Department of Genetics and Development, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
22
|
Moura RA, Fonseca JE. JAK Inhibitors and Modulation of B Cell Immune Responses in Rheumatoid Arthritis. Front Med (Lausanne) 2021; 7:607725. [PMID: 33614673 PMCID: PMC7892604 DOI: 10.3389/fmed.2020.607725] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic immune-mediated inflammatory disease that can lead to joint destruction, functional disability and substantial comorbidity due to the involvement of multiple organs and systems. B cells have several important roles in RA pathogenesis, namely through autoantibody production, antigen presentation, T cell activation, cytokine release and ectopic lymphoid neogenesis. The success of B cell depletion therapy with rituximab, a monoclonal antibody directed against CD20 expressed by B cells, has further supported B cell intervention in RA development. Despite the efficacy of synthetic and biologic disease modifying anti-rheumatic drugs (DMARDs) in the treatment of RA, few patients reach sustained remission and refractory disease is a concern that needs critical evaluation and close monitoring. Janus kinase (JAK) inhibitors or JAKi are a new class of oral medications recently approved for the treatment of RA. JAK inhibitors suppress the activity of one or more of the JAK family of tyrosine kinases, thus interfering with the JAK-Signal Transducer and Activator of Transcription (STAT) signaling pathway. To date, there are five JAK inhibitors (tofacitinib, baricitinib, upadacitinib, peficitinib and filgotinib) approved in the USA, Europe and/ or Japan for RA treatment. Evidence from the literature indicates that JAK inhibitors interfere with B cell functions. In this review, the main results obtained in clinical trials, pharmacokinetic, in vitro and in vivo studies concerning the effects of JAK inhibitors on B cell immune responses in RA are summarized.
Collapse
Affiliation(s)
- Rita A Moura
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - João Eurico Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisbon Academic Medical Centre, Lisbon, Portugal
| |
Collapse
|
23
|
Development of JAK inhibitors for the treatment of immune-mediated diseases: kinase-targeted inhibitors and pseudokinase-targeted inhibitors. Arch Pharm Res 2020; 43:1173-1186. [PMID: 33161563 DOI: 10.1007/s12272-020-01282-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/29/2020] [Indexed: 12/16/2022]
Abstract
JAKs are a family of intracellular tyrosine kinases consisting of four members, JAK1, JAK2, JAK3, and TYK2. They are key components of the JAK-STAT pathway that transmit signals of many cytokines involved in the pathogenesis of numerous immune-mediated diseases and have been major molecular targets in developing new drugs for the treatment of such diseases. Some small-molecule inhibitors of JAKs have been approved by the FDA for rheumatoid arthritis, psoriatic arthritis, and inflammatory bowel disease. Now, newer JAK inhibitors with isoform-selectivity among the four different JAKs are being developed, with the aim of improving clinical outcomes compared with earlier developed drugs with pan-JAK inhibition. Most of these selective inhibitors target the kinase domains of JAKs, functioning through the traditional inhibition mode of kinases; but recently those that target their pseudokinase domains, allosterically inhibiting the enzymes, have been under development. In this review, key characteristics, efficacy, and safety of FDA-approved and representative drugs in late stages of development are briefly described in order to provide clinical implications with respect to JAK inhibitor selectivity and future development perspectives. The recent development of pseudokinase-targeted inhibitors of JAKs is also included.
Collapse
|
24
|
El Jammal T, Sève P, Gerfaud-Valentin M, Jamilloux Y. State of the art: approved and emerging JAK inhibitors for rheumatoid arthritis. Expert Opin Pharmacother 2020; 22:205-218. [PMID: 32967471 DOI: 10.1080/14656566.2020.1822325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is the most common autoimmune inflammatory arthritis in adults. In the past decade, many treatments have emerged to expand the therapeutic armamentarium of rheumatologists. Among emerging treatments, Janus Kinase inhibitors (JAKi) are promising in treating RA and several other inflammatory conditions, such as psoriatic arthritis (PsA). The JAK/STAT signaling pathway is located downstream certain cytokines receptors that are known to be involved in RA pathogenesis. So far, three JAKi are approved for the treatment of RA, while other JAKi, are under investigation. AREAS COVERED Herein, the authors review those JAKi approved and emerging for the treatment of RA and provide their expert perspectives on the subject area. EXPERT OPINION JAKi represent an interesting alternative to other DMARDs when MTX has failed. Long-term extension studies are still ongoing, but one can assume that most of the major safety concerns have already come out. Switching from one JAKi to another DMARD has been little studied, but in such cases, preferring a treatment which does not interfere with the JAK/STAT pathway seems to be a reasonable choice.
Collapse
Affiliation(s)
- Thomas El Jammal
- Department of Internal Medicine, Lyon University Hospital , Lyon, France
| | - Pascal Sève
- Department of Internal Medicine, Lyon University Hospital , Lyon, France
| | | | - Yvan Jamilloux
- Department of Internal Medicine, Lyon University Hospital , Lyon, France
| |
Collapse
|
25
|
Liu Y, Cook C, Sedgewick AJ, Zhang S, Fassett MS, Ricardo-Gonzalez RR, Harirchian P, Kashem SW, Hanakawa S, Leistico JR, North JP, Taylor MA, Zhang W, Man MQ, Charruyer A, Beliakova-Bethell N, Benz SC, Ghadially R, Mauro TM, Kaplan DH, Kabashima K, Choi J, Song JS, Cho RJ, Cheng JB. Single-Cell Profiling Reveals Divergent, Globally Patterned Immune Responses in Murine Skin Inflammation. iScience 2020; 23:101582. [PMID: 33205009 PMCID: PMC7648132 DOI: 10.1016/j.isci.2020.101582] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/12/2020] [Accepted: 09/16/2020] [Indexed: 01/01/2023] Open
Abstract
Inflammatory response heterogeneity has impeded high-resolution dissection of diverse immune cell populations during activation. We characterize mouse cutaneous immune cells by single-cell RNA sequencing, after inducing inflammation using imiquimod and oxazolone dermatitis models. We identify 13 CD45+ subpopulations, which broadly represent most functionally characterized immune cell types. Oxazolone pervasively upregulates Jak2/Stat3 expression across T cells and antigen-presenting cells (APCs). Oxazolone also induces Il4/Il13 expression in newly infiltrating basophils, and Il4ra and Ccl24, most prominently in APCs. In contrast, imiquimod broadly upregulates Il17/Il22 and Ccl4/Ccl5. A comparative analysis of single-cell inflammatory transcriptional responses reveals that APC response to oxazolone is tightly restricted by cell identity, whereas imiquimod enforces shared programs on multiple APC populations in parallel. These global molecular patterns not only contrast immune responses on a systems level but also suggest that the mechanisms of new sources of inflammation can eventually be deduced by comparison to known signatures. Oxazolone pervasively upregulates Jak2/Stat3 expression across T cells and APCs Il4/Il13 induction in skin by oxazolone is dominated by infiltrating basophils Imiquimod broadly increases Il17/Il22 and Ccl4/Ccl5, extending to non-T cells Oxazolone induces more highly compartmentalized immune cell responses than imiquimod
Collapse
Affiliation(s)
- Yale Liu
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA, USA
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, ShaanXi, China
| | - Christopher Cook
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA, USA
| | | | - Shuyi Zhang
- Department of Physics, Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Marlys S. Fassett
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
- Department of Immunology and Microbiology, University of California, San Francisco, San Francisco, CA, USA
| | - Roberto R. Ricardo-Gonzalez
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
- Department of Immunology and Microbiology, University of California, San Francisco, San Francisco, CA, USA
| | - Paymann Harirchian
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA, USA
| | - Sakeen W. Kashem
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Sho Hanakawa
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Jacob R. Leistico
- Department of Physics, Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Jeffrey P. North
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Mark A. Taylor
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Wei Zhang
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA, USA
| | - Mao-Qiang Man
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA, USA
| | - Alexandra Charruyer
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA, USA
| | - Nadejda Beliakova-Bethell
- Department of Medicine, University of California San Diego, La Jolla, CA 92093-0679, USA
- Veterans Affairs Medical Center, San Diego, CA, USA
| | | | - Ruby Ghadially
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA, USA
| | - Theodora M. Mauro
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA, USA
| | - Daniel H. Kaplan
- Departments of Dermatology and Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Singapore Immunology Network (SIgN) and Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Jaehyuk Choi
- Department of Dermatology, Northwestern School of Medicine, Chicago, IL, USA
| | - Jun S. Song
- Department of Physics, Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Raymond J. Cho
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
- Corresponding author
| | - Jeffrey B. Cheng
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA, USA
- Corresponding author
| |
Collapse
|
26
|
Wang Q, Zhou X, Yang L, Zhao Y, Chew Z, Xiao J, Liu C, Zheng X, Zheng Y, Shi Q, Liang Q, Wang Y, Wang H. The Natural Compound Notopterol Binds and Targets JAK2/3 to Ameliorate Inflammation and Arthritis. Cell Rep 2020; 32:108158. [PMID: 32937124 DOI: 10.1016/j.celrep.2020.108158] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/18/2020] [Accepted: 08/26/2020] [Indexed: 01/06/2023] Open
Abstract
The traditional Chinese medicinal herb Notopterygium incisum Ting ex H.T. Chang has anti-rheumatism activity, and a mass spectrometry assay of patients' serum after administration of the herb revealed that notopterol is the most abundant component enriched. However, the functions of notopterol and its molecular target in rheumatoid arthritis (RA) treatment remain unknown. Here, we show in different RA mouse strains that both oral and intraperitoneal administration of notopterol result in significant therapeutic effects. Mechanistically, notopterol directly binds Janus kinase (JAK)2 and JAK3 kinase domains to inhibit JAK/signal transducers and activators of transcription (JAK-STAT) activation, leading to reduced production of inflammatory cytokines and chemokines. Critically, combination therapy using both notopterol and tumor necrosis factor (TNF) blocker results in enhanced therapeutic effects compared to using TNF blocker alone. We demonstrate that notopterol ameliorates RA pathology by targeting JAK-STAT signaling, raising the possibility that notopterol could be effective in treating other diseases characterized by aberrant JAK-STAT signaling pathway.
Collapse
Affiliation(s)
- Qiong Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; Cancer Center, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China
| | - Long Yang
- Department of Rehabilitation Medicine, Shanghai Eighth People's Hospital, Shanghai 200235, China
| | - Yongjian Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Zhihuan Chew
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jun Xiao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Chang Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; Cancer Center, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China
| | - Xin Zheng
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuxiao Zheng
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Qi Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Hongyan Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| |
Collapse
|
27
|
Singh S, Singh S. JAK-STAT inhibitors: Immersing therapeutic approach for management of rheumatoid arthritis. Int Immunopharmacol 2020; 86:106731. [PMID: 32590315 DOI: 10.1016/j.intimp.2020.106731] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 12/16/2022]
Abstract
Rheumatoid arthritis is a world leading cause of musculoskeletal disease. With the introduction of biological agents as treatment alternatives the clinical possibilities have grown exponentially. Currently most common Disease-modifying anti-rheumatic drugs (DMARDs) treatment option involves intravenous or subcutaneous injection, and some patients struggle to respond to DMARDs or lose their primary reaction. An oral drug formulation with lowered costs of manufacturing and flexibility for healthcare workers to preferably perform treatment will result in decreased healthcare expenditures and increased medication compliance. The JAK-STAT inhibitors, a new class of small molecules drugs, fulfills these criteria and has recently shown efficacy in rheumatoid arthritis. Here we give a summary of how JAK-STAT inhibitors function and a detailed review of current clinical trials. Convincing clinical results suggest that therapeutic inhibition of the JAK proteins can effectively modulate a complex cytokine-driven inflammation.
Collapse
Affiliation(s)
- Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, Hajipur, Bihar, India.
| | - Shantanu Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, Hajipur, Bihar, India
| |
Collapse
|
28
|
Burja B, Mertelj T, Frank-Bertoncelj M. Hi- JAKi-ng Synovial Fibroblasts in Inflammatory Arthritis With JAK Inhibitors. Front Med (Lausanne) 2020; 7:124. [PMID: 32432116 PMCID: PMC7214667 DOI: 10.3389/fmed.2020.00124] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/20/2020] [Indexed: 12/23/2022] Open
Abstract
The Janus kinase (JAK)-Signal transducer and activator of transcription (STAT) pathway is one of the central signaling hubs in inflammatory, immune and cancer cells. Inhibiting the JAK-STAT pathway with JAK inhibitors (jakinibs) constitutes an important therapeutic strategy in cancer and chronic inflammatory diseases like rheumatoid arthritis (RA). FDA has approved different jakinibs for the treatment of RA, including tofacitinib, baricitinib and upadacitinib, and several jakinibs are being tested in clinical trials. Here, we reviewed published studies of jakinib effects on resolving synovial pathology in inflammatory arthritis. We discussed the results of jakinibs on structural joint damage in clinical trials and explored the effects of jakinibs across different in vitro, ex vivo, and in vivo synovial experimental models. We delved rigorously into experimental designs of in vitro fibroblast studies, deconvoluted jakinib efficacy in synovial fibroblasts across diverse experimental conditions and discussed their translatability in vivo. Synovial fibroblasts can readily activate the JAK-STAT signaling pathway in response to cytokine stimulation. We highlighted rather limited effects of jakinibs on the in vitro cultured synovial fibroblasts and inferred that direct and indirect (immune cell-dependent) actions of jakinibs are required to curb the fibroblast pathology in vivo. These actions have not been mimicked optimally in current in vitro experimental designs, where inflammatory stimuli do not naturally clear out with treatment as they do in vivo. While summarizing the broad knowledge of synovial jakinib effects, our review uniquely challenges future study designs to better mimick the jakinib actions in broader cell communities, as occurring in vivo in the inflamed synovium. This can deepen our understanding of collective synovial activities of jakinibs and their therapeutic limitations, thereby fostering jakinib development in arthritis.
Collapse
Affiliation(s)
- Blaž Burja
- Center of Experimental Rheumatology, University Hospital Zurich, Schlieren, Switzerland.,Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tonja Mertelj
- Center of Experimental Rheumatology, University Hospital Zurich, Schlieren, Switzerland
| | | |
Collapse
|
29
|
Xu P, Shen P, Yu B, Xu X, Ge R, Cheng X, Chen Q, Bian J, Li Z, Wang J. Janus kinases (JAKs): The efficient therapeutic targets for autoimmune diseases and myeloproliferative disorders. Eur J Med Chem 2020; 192:112155. [PMID: 32120325 DOI: 10.1016/j.ejmech.2020.112155] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/16/2020] [Accepted: 02/16/2020] [Indexed: 02/06/2023]
Abstract
The Janus kinases or JAKs are a family of intracellular tyrosine kinases that play an essential role in the signaling of numerous cytokines that have been implicated in the pathogenesis of autoimmune diseases and myeloproliferative disorders. JAKs are activated upon ligand induced receptor homo- or heterodimerization, which results in the immediate phosphorylation of tyrosine residues and the phosphotyrosines then serve as docking sites for cytoplasmic signal transducer and activator of transcription (STAT) proteins which become phosphorylated by the JAKs upon recruitment to the receptor complex. The phosphorylated STAT proteins dimerize and travel to the cellular nucleus, where they act as transcription factors. Interfering in the JAK-STAT pathway has yielded the only approved small molecule kinase inhibitors for immunological indications. Numerous medicinal chemistry studies are currently aimed at the design of novel and potent inhibitors for JAKs. Additionally, whether the second-generation inhibitors which possessed selectivity for JAKs are more efficient are under research. This Perspective summarizes the progress in the discovery and development of JAKs inhibitors, including the potential binding site and approaches for identifying small-molecule inhibitors, as well as future therapeutic perspectives in autoimmune diseases and myeloproliferative disorders are also put forward in order to provide reference and rational for the drug discovery of novel and potent JAKs inhibitors.
Collapse
Affiliation(s)
- Pengfei Xu
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Pei Shen
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Bin Yu
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Xi Xu
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Raoling Ge
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming, 650000, China
| | - Xinying Cheng
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Qiuyu Chen
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Jinlei Bian
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 21009, China
| | - Zhiyu Li
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 21009, China.
| | - JuBo Wang
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 21009, China.
| |
Collapse
|
30
|
Wu J, Zhu Z, Yu Q, Ding C. Tyrosine kinase inhibitors for the treatment of rheumatoid arthritis: phase I to Ⅱ clinical trials. Expert Opin Investig Drugs 2019; 28:1113-1123. [PMID: 31738612 DOI: 10.1080/13543784.2019.1692812] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Introduction: Rheumatoid arthritis (RA) is a chronic, refractory disorder caused by autoimmunity in the synovial joints. Disease-modifying anti-rheumatic drugs (DMARDs) and biologicals offer remission in only two-thirds of RA patients within 3 months, hence new therapeutic approaches are necessary. Tyrosine kinase inhibitors (TKIs) are newly developed small molecule drugs which have demonstrated encouraging results in this disease.Areas covered: The key findings from phase I and II clinical trials that have investigated the use of novel TKIs in the treatment of RA are discussed. We examined the literature published between January 2014 to January 2019 using electronic databases including PubMed, Web of Science, Medline, Embase, and Google Scholar. Additional information about phase I and II trials on the ClinicalTrial.gov website up to January 2019 was also retrieved.Expert opinion: JAK inhibitors are promising drugs with sound efficacy and acceptable safety and may be beneficial to patients who do not respond to DMARDs and biologicals. The response rates among RA patients to TKIs are diverse; genetic and environmental factors may be involved in the varying responses which are closely related to the pathogenesis of RA. Future studies may reveal the underlying mechanisms of resistance and non-response.
Collapse
Affiliation(s)
- Jing Wu
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhaohua Zhu
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qinghong Yu
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Changhai Ding
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| |
Collapse
|
31
|
Mei H, Han J, Klika KD, Izawa K, Sato T, Meanwell NA, Soloshonok VA. Applications of fluorine-containing amino acids for drug design. Eur J Med Chem 2019; 186:111826. [PMID: 31740056 DOI: 10.1016/j.ejmech.2019.111826] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/21/2019] [Accepted: 10/26/2019] [Indexed: 01/26/2023]
Abstract
Fluorine-containing amino acids are becoming increasingly prominent in new drugs due to two general trends in the modern pharmaceutical industry. Firstly, the growing acceptance of peptides and modified peptides as drugs; and secondly, fluorine editing has become a prevalent protocol in drug-candidate optimization. Accordingly, fluorine-containing amino acids represent one of the more promising and rapidly developing areas of research in organic, bio-organic and medicinal chemistry. The goal of this Review article is to highlight the current state-of-the-art in this area by profiling 42 selected compounds that combine fluorine and amino acid structural elements. The compounds under discussion represent pharmaceutical drugs currently on the market, or in clinical trials as well as examples of drug-candidates that although withdrawn from development had a significant impact on the progress of medicinal chemistry and/or provided a deeper understanding of the nature and mechanism of biological action. For each compound, we present features of biological activity, a brief history of the design principles and the development of the synthetic approach, focusing on the source of tailor-made amino acid structures and fluorination methods. General aspects of the medicinal chemistry of fluorine-containing amino acids and synthetic methodology are briefly discussed.
Collapse
Affiliation(s)
- Haibo Mei
- College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Jianlin Han
- College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Karel D Klika
- Molecular Structure Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | - Kunisuke Izawa
- Hamari Chemicals Ltd., 1-4-29 Kunijima, Higashi-Yodogawa-ku, Osaka, 533-0024, Japan.
| | - Tatsunori Sato
- Hamari Chemicals Ltd., 1-4-29 Kunijima, Higashi-Yodogawa-ku, Osaka, 533-0024, Japan
| | - Nicholas A Meanwell
- Department of Discovery Chemistry, Bristol-Myers Squibb Research and Development, PO Box 4000, Princeton, NJ, 08543-4000, United States.
| | - Vadim A Soloshonok
- Department of Organic Chemistry I, University of the Basque Country UPV/EHU, Paseo Manuel Lardizábal 3, 20018, San Sebastián, Spain; IKERBASQUE, Basque Foundation for Science, María Díaz de Haro 3, Plaza Bizkaia, 48013, Bilbao, Spain.
| |
Collapse
|
32
|
Sonthalia S, Aggarwal P. Oral Tofacitinib: Contemporary Appraisal of Its Role in Dermatology. Indian Dermatol Online J 2019; 10:503-518. [PMID: 31544068 PMCID: PMC6743407 DOI: 10.4103/idoj.idoj_474_18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Tofacitinib, an oral Janus kinase inhibitor (Jakinib), is an emerging treatment modality whose well-established efficacy in systemic inflammatory diseases is now being actively explored for cutaneous disorders (arising due to the patient's dysimmune responses) that are not responding to and/or sustaining intolerable adverse effects with the classical immunosuppressives and other targeted therapies such as the biologics. The most common dermatoses for which oral as well as topical Jakinibs such as tofacitinib have been evaluated and are being used albeit as an off-label indication include psoriasis, psoriatic arthritis, alopecia areata, vitiligo, and atopic dermatitis. This article provides a succinct review on the current status of oral tofacitinib in dermatology through literature search of PubMed database and stresses on the need for further evidence generation to define the drug's place in the therapeutic arsenal of dysimmune cutaneous disorders.
Collapse
Affiliation(s)
- Sidharth Sonthalia
- Medical Director and Senior Consultant Dermatologist, Department of Dermatology and Dermatosurgery, Skinnocence: The Skin Clinic and Research Center, Gurugram, Haryana, India
| | - Parul Aggarwal
- Consultant Dermatologist, Department of Dermatology and STD, Skinalive Clinic, New Delhi, India
| |
Collapse
|
33
|
Alunno A, Padjen I, Fanouriakis A, Boumpas DT. Pathogenic and Therapeutic Relevance of JAK/STAT Signaling in Systemic Lupus Erythematosus: Integration of Distinct Inflammatory Pathways and the Prospect of Their Inhibition with an Oral Agent. Cells 2019; 8:cells8080898. [PMID: 31443172 PMCID: PMC6721755 DOI: 10.3390/cells8080898] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 12/21/2022] Open
Abstract
Four Janus kinases (JAKs) (JAK1, JAK2, JAK3, TYK2) and seven signal transducers and activators of transcription (STATs) (STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B, STAT6) mediate the signal transduction of more than 50 cytokines and growth factors in many different cell types. Located intracellularly and downstream of cytokine receptors, JAKs integrate and balance the actions of various signaling pathways. With distinct panels of STAT-sensitive genes in different tissues, this highly heterogeneous system has broad in vivo functions playing a crucial role in the immune system. Thus, the JAK/STAT pathway is critical for resisting infection, maintaining immune tolerance, and enforcing barrier functions and immune surveillance against cancer. Breakdowns of this system and/or increased signal transduction may lead to autoimmunity and other diseases. Accordingly, the recent development and approval of the first small synthetic molecules targeting JAK molecules have opened new therapeutic avenues of potentially broad therapeutic relevance. Extensive data are now available regarding the JAK/STAT pathway in rheumatoid arthritis. Dysregulation of the cytokines is also a hallmark of systemic lupus erythematosus (SLE), and targeting the JAK/STAT proteins allows simultaneous suppression of multiple cytokines. Evidence from in vitro studies and animal models supports a pivotal role also in the pathogenesis of cutaneous lupus and SLE. This has important therapeutic implications, given the current paucity of targeted therapies especially in the latter. Herein, we summarize the currently available literature in experimental SLE, which has led to the recent promising Phase II clinical trial of a JAK inhibitor.
Collapse
Affiliation(s)
- Alessia Alunno
- Rheumatology Unit, Department of Medicine, University of Perugia, Ospedale S.M. della Misericordia, Edificio C, 5° piano, Piazzale Menghini 1, 06129 S. Andrea delle Fratte, Perugia, Italy.
| | - Ivan Padjen
- Division of Clinical Immunology and Rheumatology, Department of Internal Medicine, University Hospital Center Zagreb and University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Antonis Fanouriakis
- Rheumatology and Clinical Immunology Unit, 4th Department of Internal Medicine, "Attikon" University Hospital, 12462 Athens, Greece
- Department of Rheumatology, "Asklepieion" General Hospital, 16673 Athens, Greece
| | - Dimitrios T Boumpas
- Rheumatology and Clinical Immunology Unit, 4th Department of Internal Medicine, "Attikon" University Hospital, 12462 Athens, Greece
- Laboratory of Autoimmunity and Inflammation, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
- Joint Academic Rheumatology Program, Medical School, National and Kapodestrian University of Athens, Athens, Greece and Medical School, University of Cyprus, 1678 Nicosia, Cyprus
| |
Collapse
|
34
|
Vyas SP, Hansda AK, Goswami R. Rheumatoid arthritis: ‘melting pot’ of T helper subsets. Int Rev Immunol 2019; 38:212-231. [PMID: 31155981 DOI: 10.1080/08830185.2019.1621865] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
35
|
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
|
36
|
T Virtanen A, Haikarainen T, Raivola J, Silvennoinen O. Selective JAKinibs: Prospects in Inflammatory and Autoimmune Diseases. BioDrugs 2019; 33:15-32. [PMID: 30701418 PMCID: PMC6373396 DOI: 10.1007/s40259-019-00333-w] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cytokines, many of which signal through the JAK-STAT (Janus kinase-Signal Transducers and Activators of Transcription) pathway, play a central role in the pathogenesis of inflammatory and autoimmune diseases. Currently three JAK inhibitors have been approved for clinical use in USA and/or Europe: tofacitinib for rheumatoid arthritis, psoriatic arthritis and ulcerative colitis, baricitinib for rheumatoid arthritis, and ruxolitinib for myeloproliferative neoplasms. The clinical JAK inhibitors target multiple JAKs at high potency and current research has focused on more selective JAK inhibitors, almost a dozen of which currently are being evaluated in clinical trials. In this narrative review, we summarize the status of the pan-JAK and selective JAK inhibitors approved or in clinical trials, and discuss the rationale for selective targeting of JAKs in inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Anniina T Virtanen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland.
| | - Teemu Haikarainen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Juuli Raivola
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Olli Silvennoinen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland.
- Fimlab Laboratories, 33520, Tampere, Finland.
- Institute of Biotechnology, University of Helsinki, P.O. Box 56, (Viikinkaari 5), 00014, Helsinki, Finland.
| |
Collapse
|
37
|
Westhovens R. Clinical efficacy of new JAK inhibitors under development. Just more of the same? Rheumatology (Oxford) 2019; 58:i27-i33. [PMID: 30806706 PMCID: PMC6390876 DOI: 10.1093/rheumatology/key256] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 07/17/2018] [Indexed: 01/07/2023] Open
Abstract
Janus kinase inhibition is promising in the treatment of RA, with already two oral drugs marketed. New compounds are under investigation that are more selective for Janus kinase 1 or Janus kinase 3. Phase II results for filgotinib, upadacitinib, peficitinib and decernotinib are reviewed showing almost consistently a fast dose-dependent clinical improvement similar to already approved drugs tofacitinib and baricitinib. I will reflect on the most frequently reported dose-dependent adverse events and laboratory changes. Some are similar for all drugs of this class, some are more specific for a certain drug, but all may influence future treatment effectiveness in daily practice. This implies the need for a critical evaluation of phase III trials, and eventually trials specifically powered for conclusions on the safety profile and registries once these drugs become marketed. These innovative drugs also need head-to-head trials versus biologics or in-class as well as specific strategy studies to determine their optimal future use.
Collapse
Affiliation(s)
- Rene Westhovens
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
38
|
Malemud CJ. The role of the JAK/STAT signal pathway in rheumatoid arthritis. Ther Adv Musculoskelet Dis 2018; 10:117-127. [PMID: 29942363 PMCID: PMC6009092 DOI: 10.1177/1759720x18776224] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/20/2018] [Indexed: 12/30/2022] Open
Abstract
Proinflammatory cytokine activation of the Janus kinase/signal transducers and activators of transcription (JAK/STAT) signal transduction pathway is a critical event in the pathogenesis and progression of rheumatoid arthritis. Under normal conditions, JAK/STAT signaling reflects the influence of negative regulators of JAK/STAT, exemplified by the suppressor of cytokine signaling and protein inhibitor of activated STAT. However, in rheumatoid arthritis (RA) both of these regulators are dysfunctional. Thus, continuous activation of JAK/STAT signaling in RA synovial joints results in the elevated level of matrix metalloproteinase gene expression, increased frequency of apoptotic chondrocytes and most prominently 'apoptosis resistance' in the inflamed synovial tissue. Tofacitinib, a JAK small molecule inhibitor, with selectivity for JAK2/JAK3 was approved by the United States Food and Drug Administration (US FDA) for the therapy of RA. Importantly, tofacitinib has demonstrated significant clinical efficacy for RA in the post-US FDA-approval surveillance period. Of note, the success of tofacitinib has spurred the development of JAK1, JAK2 and other JAK3-selective small molecule inhibitors, some of which have also entered the clinical setting, whereas other JAK inhibitors are currently being evaluated in RA clinical trials.
Collapse
|
39
|
Laribi K, Lemaire P, Sandrini J, Baugier de Materre A. Advances in the understanding and management of T-cell prolymphocytic leukemia. Oncotarget 2017; 8:104664-104686. [PMID: 29262669 PMCID: PMC5732835 DOI: 10.18632/oncotarget.22272] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 08/27/2017] [Indexed: 12/02/2022] Open
Abstract
T-prolymphocytic leukemia (T-PLL) is a rare T-cell neoplasm with an aggressive clinical course. Leukemic T-cells exhibit a post-thymic T-cell phenotype (Tdt-, CD1a-, CD5+, CD2+ and CD7+) and are generally CD4+/CD8-, but CD4+/CD8+ or CD8+/CD4- T-PLL have also been reported. The hallmark of T-PLL is the rearrangement of chromosome 14 involving genes for the subunits of the T-cell receptor (TCR) complex, leading to overexpression of the proto-oncogene TCL1. In addition, molecular analysis shows that T-PLL exhibits substantial mutational activation of the IL2RG-JAK1-JAK3-, STAT5B axis. T-PLL patients have a poor prognosis, due to a poor response to conventional chemotherapy. Monoclonal antibody therapy with antiCD52-alemtuzumab has considerably improved outcomes, but the responses to treatment are transient; hence, patients who achieve a response to therapy are considered for stem cell transplantation (SCT). This combined approach has extended the median survival to four years or more. Nevertheless, new approaches using well-tolerated therapies that target growth and survival signals are needed for most patients unable to receive intensive chemotherapy.
Collapse
Affiliation(s)
- Kamel Laribi
- Department of Hematology, Centre Hospitalier du Mans, Le Mans, France
| | - Pierre Lemaire
- Laboratory of Biology and Hematology, Centre Hospitalier du Mans, Le Mans, France
| | - Jeremy Sandrini
- Laboratory of Anatomopathology, Centre Hospitalier du Mans, Le Mans, France
| | | |
Collapse
|
40
|
DeMars KM, Pacheco SC, Yang C, Siwarski DM, Candelario-Jalil E. Selective Inhibition of Janus Kinase 3 Has No Impact on Infarct Size or Neurobehavioral Outcomes in Permanent Ischemic Stroke in Mice. Front Neurol 2017; 8:363. [PMID: 28790974 PMCID: PMC5524742 DOI: 10.3389/fneur.2017.00363] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/10/2017] [Indexed: 11/20/2022] Open
Abstract
Janus kinase 3 (JAK3) is associated with the common gamma chain of several interleukin (IL) receptors essential to inflammatory signaling. To study the potential role of JAK3 in stroke-induced neuroinflammation, we subjected mice to permanent middle cerebral artery occlusion and investigated the effects of JAK3 inhibition with decernotinib (VX-509) on infarct size, behavior, and levels of several inflammatory mediators. Results from our double immunofluorescence staining showed JAK3 expression on neurons, endothelial cells, and microglia/macrophages in the ischemic mouse brain (n = 3). We found for the first time that total and phosphorylated/activated JAK3 are dramatically increased after stroke in the ipsilateral hemisphere (**P < 0.01; n = 5–13/group) in addition to increased IL-21 expression after stroke (**P < 0.01; n = 5–7/group). However, inhibition of JAK3 confirmed by reduced phosphorylation of its activation loop at tyrosine residues 980/981 does not reduce infarct volume measured at 48 h after stroke (n = 6–10/group) nor does it alter behavioral outcomes sensitive to neurological deficits or stroke-induced neuroinflammatory response (n = 9–10/group). These results do not support a detrimental role for JAK3 in acute neuroinflammation following permanent focal cerebral ischemia. The functional role of increased JAK3 activation after stroke remains to be further investigated.
Collapse
Affiliation(s)
- Kelly M DeMars
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Sean C Pacheco
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - David M Siwarski
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
41
|
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
|
42
|
Banerjee S, Biehl A, Gadina M, Hasni S, Schwartz DM. JAK-STAT Signaling as a Target for Inflammatory and Autoimmune Diseases: Current and Future Prospects. Drugs 2017; 77:521-546. [PMID: 28255960 PMCID: PMC7102286 DOI: 10.1007/s40265-017-0701-9] [Citation(s) in RCA: 777] [Impact Index Per Article: 97.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The Janus kinase/signal transduction and activator of transcription (JAK-STAT) signaling pathway is implicated in the pathogenesis of inflammatory and autoimmune diseases including rheumatoid arthritis, psoriasis, and inflammatory bowel disease. Many cytokines involved in the pathogenesis of autoimmune and inflammatory diseases use JAKs and STATs to transduce intracellular signals. Mutations in JAK and STAT genes cause a number of immunodeficiency syndromes, and polymorphisms in these genes are associated with autoimmune diseases. The success of small-molecule JAK inhibitors (Jakinibs) in the treatment of rheumatologic disease demonstrates that intracellular signaling pathways can be targeted therapeutically to treat autoimmunity. Tofacitinib, the first rheumatologic Jakinib, is US Food and Drug Administration (FDA) approved for rheumatoid arthritis and is currently under investigation for other autoimmune diseases. Many other Jakinibs are in preclinical development or in various phases of clinical trials. This review describes the JAK-STAT pathway, outlines its role in autoimmunity, and explains the rationale/pre-clinical evidence for targeting JAK-STAT signaling. The safety and clinical efficacy of the Jakinibs are reviewed, starting with the FDA-approved Jakinib tofacitinib, and continuing on to next-generation Jakinibs. Recent and ongoing studies are emphasized, with a focus on emerging indications for JAK inhibition and novel mechanisms of JAK-STAT signaling blockade.
Collapse
Affiliation(s)
- Shubhasree Banerjee
- Rheumatology Fellowship and Training Branch, National Institute of Arthritis Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | - Ann Biehl
- Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Massimo Gadina
- Translational Immunology Section, National Institute of Arthritis Musculoskeletal and Skin diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sarfaraz Hasni
- Lupus Clinical Research Program, National Institute of Arthritis Musculoskeletal and Skin diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniella M Schwartz
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis Musculoskeletal and Skin diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
43
|
Lin YY, Jean YH, Lee HP, Lin SC, Pan CY, Chen WF, Wu SF, Su JH, Tsui KH, Sheu JH, Sung PJ, Wen ZH. Excavatolide B Attenuates Rheumatoid Arthritis through the Inhibition of Osteoclastogenesis. Mar Drugs 2017; 15:md15010009. [PMID: 28067799 PMCID: PMC5295229 DOI: 10.3390/md15010009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/17/2016] [Accepted: 12/26/2016] [Indexed: 02/08/2023] Open
Abstract
Osteoclasts are multinucleated giant cells of macrophage/monocyte lineage, and cell differentiation with the upregulation of osteoclast-related proteins is believed to play a major role in the destruction of the joints in the course of rheumatoid arthritis (RA). Pro-inflammatory cytokines, such as interleukin-17A (IL-17A) and macrophage colony-stimulating factor (M-CSF), can be overexpressed in RA and lead to osteoclastogenesis. In a previous study, we found that cultured-type soft coral-derived excavatolide B (Exc-B) exhibited anti-inflammatory properties. In the present study, we thus aimed to evaluate the anti-arthritic activity of Exc-B in in vitro and in vivo models. The results demonstrated that Exc-B inhibits LPS-induced multinucleated cell and actin ring formation, as well as TRAP, MMP-9, and cathepsin K expression. Additionally, Exc-B significantly attenuated the characteristics of RA in adjuvant (AIA) and type II collagen-induced arthritis (CIA) in rats. Moreover, Exc-B improved histopathological features, and reduced the number of TRAP-positive multinucleated cells in the in vivo AIA and CIA models. Immunohistochemical analysis showed that Exc-B attenuated the protein expression of cathepsin K, MMP-2, MMP-9, CD11b, and NFATc1 in ankle tissues of AIA and CIA rats. Level of interleukin-17A and macrophage colony-stimulating factor were also decreased by Exc-B. These findings strongly suggest that Exc-B could be of potential use as a therapeutic agent by inhibiting osteoclast differentiation in arthritis. Moreover, this study also illustrates the use of the anti-inflammatory marine compound, Exc-B, as a potential therapeutic strategy for RA.
Collapse
Affiliation(s)
- Yen-You Lin
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No.70, Lianhai Road, Gushan District, Kaohsiung 80424, Taiwan.
| | - Yen-Hsuan Jean
- Department of Orthopaedic Surgery, Ping-Tung Christian Hospital, No.60, Dalian Road, Pingtung 90059, Taiwan.
| | - Hsin-Pai Lee
- Department of Orthopaedic Surgery, Ping-Tung Christian Hospital, No.60, Dalian Road, Pingtung 90059, Taiwan.
| | - Sung-Chun Lin
- Department of Orthopaedic Surgery, Ping-Tung Christian Hospital, No.60, Dalian Road, Pingtung 90059, Taiwan.
| | - Chieh-Yu Pan
- Department and Graduate Institute of Aquaculture, National Kaohsiung Marine University, No.142, Haizhuan Road, Nanzi District, Kaohsiung 81157, Taiwan.
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No.70, Lianhai Road, Gushan District, Kaohsiung 80424, Taiwan.
- Department of Neurosurgery, Chang Gung Memorial Hospital-Kaohsiung Medical Center and Chang Gung University College of Medicine, No.123, Dapi Road, Niaosong District, Kaohsiung 83301, Taiwan.
- Department of Neurosurgery, Xiamen Chang Gung Memorial Hospital, No.123, Xiafei Road, Haicang District, Xiamen 361000, China.
| | - Shu-Fen Wu
- Department of Life Science, Institute of Molecular Biology, National Chung-Cheng University, No.168, Sec. 1, University Road, Min-Hsiung, Chia-yi 62102, Taiwan.
| | - Jui-Hsin Su
- Taiwan Coral Research Center, National Museum of Marine Biology & Aquarium, No.2 Houwan Road, Checheng, Pingtung 94450, Taiwan.
- Graduate Institute of Marine Biotechnology, National Dong Hwa University, No.2 Houwan Road, Checheng, Pingtung 94450, Taiwan.
| | - Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Road, Zuoying District, Kaohsiung 81362, Taiwan.
- Department of Obstetrics and Gynecology and Institute of Clinical Medicine, National Yang-Ming University, No.155, Sec. 2, Linong Street, Taipei 11221, Taiwan.
- Department of Pharmacy and Graduate Institute of Pharmaceutical Technology, Tajen University, No.20, Weixin Road, Yanpu, Pingtung 90741, Taiwan.
| | - Jyh-Horng Sheu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No.70, Lianhai Road, Gushan District, Kaohsiung 80424, Taiwan.
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, No.70, Lianhai Road, Gushan District, Kaohsiung 80424, Taiwan.
| | - Ping-Jyun Sung
- Taiwan Coral Research Center, National Museum of Marine Biology & Aquarium, No.2 Houwan Road, Checheng, Pingtung 94450, Taiwan.
- Graduate Institute of Marine Biotechnology, National Dong Hwa University, No.2 Houwan Road, Checheng, Pingtung 94450, Taiwan.
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No.70, Lianhai Road, Gushan District, Kaohsiung 80424, Taiwan.
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, No.70, Lianhai Road, Gushan District, Kaohsiung 80424, Taiwan.
| |
Collapse
|
44
|
Telliez JB, Dowty ME, Wang L, Jussif J, Lin T, Li L, Moy E, Balbo P, Li W, Zhao Y, Crouse K, Dickinson C, Symanowicz P, Hegen M, Banker ME, Vincent F, Unwalla R, Liang S, Gilbert AM, Brown MF, Hayward M, Montgomery J, Yang X, Bauman J, Trujillo JI, Casimiro-Garcia A, Vajdos FF, Leung L, Geoghegan KF, Quazi A, Xuan D, Jones L, Hett E, Wright K, Clark JD, Thorarensen A. Discovery of a JAK3-Selective Inhibitor: Functional Differentiation of JAK3-Selective Inhibition over pan-JAK or JAK1-Selective Inhibition. ACS Chem Biol 2016; 11:3442-3451. [PMID: 27791347 DOI: 10.1021/acschembio.6b00677] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PF-06651600, a newly discovered potent JAK3-selective inhibitor, is highly efficacious at inhibiting γc cytokine signaling, which is dependent on both JAK1 and JAK3. PF-06651600 allowed the comparison of JAK3-selective inhibition to pan-JAK or JAK1-selective inhibition, in relevant immune cells to a level that could not be achieved previously without such potency and selectivity. In vitro, PF-06651600 inhibits Th1 and Th17 cell differentiation and function, and in vivo it reduces disease pathology in rat adjuvant-induced arthritis as well as in mouse experimental autoimmune encephalomyelitis models. Importantly, by sparing JAK1 function, PF-06651600 selectively targets γc cytokine pathways while preserving JAK1-dependent anti-inflammatory signaling such as the IL-10 suppressive functions following LPS treatment in macrophages and the suppression of TNFα and IL-1β production in IL-27-primed macrophages. Thus, JAK3-selective inhibition differentiates from pan-JAK or JAK1 inhibition in various immune cellular responses, which could potentially translate to advantageous clinical outcomes in inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Jean-Baptiste Telliez
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Martin E. Dowty
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Lu Wang
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Jason Jussif
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Tsung Lin
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Li Li
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Erick Moy
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Paul Balbo
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Wei Li
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Yajuan Zhao
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Kimberly Crouse
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Caitlyn Dickinson
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Peter Symanowicz
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Martin Hegen
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Mary Ellen Banker
- Primary Pharmacology Group, Pfizer Worldwide R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Fabien Vincent
- Primary Pharmacology Group, Pfizer Worldwide R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Ray Unwalla
- Worldwide Medicinal Chemistry, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Sidney Liang
- Worldwide Medicinal Chemistry, Pfizer Worldwide R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Adam M. Gilbert
- Worldwide Medicinal Chemistry, Pfizer Worldwide R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Matthew F. Brown
- Worldwide Medicinal Chemistry, Pfizer Worldwide R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Matthew Hayward
- Worldwide Medicinal Chemistry, Pfizer Worldwide R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Justin Montgomery
- Worldwide Medicinal Chemistry, Pfizer Worldwide R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Xin Yang
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jonathan Bauman
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - John I. Trujillo
- Worldwide Medicinal Chemistry, Pfizer Worldwide R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Agustin Casimiro-Garcia
- Worldwide Medicinal Chemistry, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Felix F. Vajdos
- Worldwide Medicinal Chemistry, Pfizer Worldwide R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Louis Leung
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kieran F. Geoghegan
- Worldwide Medicinal Chemistry, Pfizer Worldwide R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Amira Quazi
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Dejun Xuan
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Lyn Jones
- Worldwide Medicinal Chemistry, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Erik Hett
- Worldwide Medicinal Chemistry, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Katherine Wright
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - James D. Clark
- Inflammation and Immunology, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Atli Thorarensen
- Worldwide Medicinal Chemistry, Pfizer Worldwide R&D, 610 Main Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
45
|
Semerano L, Decker P, Clavel G, Boissier MC. Developments with investigational Janus kinase inhibitors for rheumatoid arthritis. Expert Opin Investig Drugs 2016; 25:1355-1359. [PMID: 27748152 DOI: 10.1080/13543784.2016.1249565] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Luca Semerano
- a Inserm UMR 1125 , Bobigny , France.,b Sorbonne Paris Cité , Université Paris 13 , Bobigny , France.,c Department of Rheumatology , Hôpitaux Universitaires Paris-Seine-Saint Denis, Assistance-Publique-Hôpitaux de Paris (APHP) , Bobigny , France
| | - Patrice Decker
- a Inserm UMR 1125 , Bobigny , France.,b Sorbonne Paris Cité , Université Paris 13 , Bobigny , France
| | - Gaëlle Clavel
- b Sorbonne Paris Cité , Université Paris 13 , Bobigny , France.,d Department of Internal Medicine , Fondation Rothschild , Paris , France
| | - Marie-Christophe Boissier
- a Inserm UMR 1125 , Bobigny , France.,b Sorbonne Paris Cité , Université Paris 13 , Bobigny , France.,c Department of Rheumatology , Hôpitaux Universitaires Paris-Seine-Saint Denis, Assistance-Publique-Hôpitaux de Paris (APHP) , Bobigny , France
| |
Collapse
|
46
|
Roskoski R. Janus kinase (JAK) inhibitors in the treatment of inflammatory and neoplastic diseases. Pharmacol Res 2016; 111:784-803. [PMID: 27473820 DOI: 10.1016/j.phrs.2016.07.038] [Citation(s) in RCA: 253] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 07/25/2016] [Accepted: 07/25/2016] [Indexed: 02/06/2023]
Abstract
The Janus kinase (JAK) family of non-receptor protein-tyrosine kinases consists of JAK1, JAK2, JAK3, and TYK2 (tyrosine kinase-2). Each of these proteins contains a JAK homology pseudokinase (JH2) domain that regulates the adjacent protein kinase domain (JH1). JAK1/2 and TYK2 are ubiquitously expressed whereas JAK3 is found predominantly in hematopoietic cells. The Janus kinase family is regulated by numerous cytokines including interleukins, interferons, and hormones such as erythropoietin, thrombopoietin, and growth hormone. Ligand binding to cytokine and hormone receptors leads to the activation of associated Janus kinases, which then mediate the phosphorylation of the receptors. The SH2 domain of STATs (signal transducers and activators of transcription) binds to the receptor phosphotyrosines thereby promoting STAT phosphorylation by the Janus kinases and consequent activation. STAT dimers are translocated to the nucleus where they participate in the regulation of the expression of thousands of proteins. JAK-STAT dysregulation results in autoimmune disorders such as rheumatoid arthritis, ulcerative colitis, and Crohn disease. JAK-STAT dysregulation also plays a role in the pathogenesis of myelofibrosis, polycythemia vera, and other myeloproliferative illnesses. An activating JAK2 V617F mutation occurs in 95% of people with polycythemia vera and in a lower percentage of people with other neoplasms. JAK1/3 signaling participates in the pathogenesis of inflammatory afflictions while JAK1/2 signaling participates in the development of several malignancies including leukemias and lymphomas as well as myeloproliferative neoplasms. Tofacitinib is a pan-JAK inhibitor that is approved by the FDA for the treatment of rheumatoid arthritis and ruxolitinib is a JAK1/2 inhibitor that is approved for the treatment of polycythemia vera and myelofibrosis.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 3754 Brevard Road, Suite 116, Box 19, Horse Shoe, NC 28742-8814, United States.
| |
Collapse
|
47
|
Gadina M, Schwartz DM, O'Shea JJ. Decernotinib: A Next-Generation Jakinib. Arthritis Rheumatol 2016; 68:31-4. [PMID: 26479275 DOI: 10.1002/art.39463] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 09/15/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Massimo Gadina
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| | - Daniella M Schwartz
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| | - John J O'Shea
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| |
Collapse
|
48
|
Novel Immunotherapeutic Avenues for Rheumatoid Arthritis. Trends Mol Med 2016; 22:214-229. [PMID: 26875450 DOI: 10.1016/j.molmed.2016.01.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 01/12/2016] [Accepted: 01/13/2016] [Indexed: 01/07/2023]
Abstract
Rheumatoid arthritis (RA) is the most common inflammatory rheumatic disease. It leads to irreversible joint damage, physical handicap, and reduced life expectancy. The past two decades have seen considerable therapeutic advances with the development of biologic treatments to block proinflammatory cytokines or modulate lymphocyte function, followed by the development of small molecules to target intracellular signaling. Nevertheless, only a minority of patients can achieve disease remission, especially long term, warranting further investigation into newer therapeutic options. Targeting single proinflammatory pathways may not be sufficient, as suggested by variable results with T helper (Th)-17-related cytokine blockade. Multilevel information from 'omics' techniques along with data from mechanistic studies might facilitate the identification of pivotal checkpoints in RA disease pathogenesis and the subsequent development of new effective treatments.
Collapse
|
49
|
Sardar S, Andersson Å. Old and new therapeutics for Rheumatoid Arthritis: in vivo models and drug development. Immunopharmacol Immunotoxicol 2016; 38:2-13. [PMID: 26769136 DOI: 10.3109/08923973.2015.1125917] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Development of novel drugs for treatment of chronic inflammatory diseases is to a large extent dependent on the availability of good experimental in vivo models in order to perform preclinical tests of new drugs and for the identification of novel drug targets. Here, we review a number of existing rodent models for Rheumatoid Arthritis in the context of how these models have been utilized for developing established therapy in Rheumatoid Arthritis and, furthermore, the present use of animal models for studies of novel drug candidates. We have studied the literature in the field for the use of in vivo models during development of anti-rheumatic drugs; from Methotrexate to various antibody treatments, to novel drugs that are, or have recently been, in clinical trials. For novel drugs, we have explored websites for clinical trials. Although a single Rheumatoid Arthritis in vivo model cannot mirror the complexity of disease development, there exist a number of good animal models for Rheumatoid Arthritis, each defining some parts in disease development, which are useful for studies of drug response. We find that many of the established drugs were not tested in in vivo models before being used in the clinic, but rather animal models have been subsequently used to find mechanisms for efficacy. Finally, we report a number of novel drugs, tested in preclinical in vivo models, presently in clinical trials.
Collapse
Affiliation(s)
- Samra Sardar
- a Department Of Drug Design and Pharmacology , Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Åsa Andersson
- a Department Of Drug Design and Pharmacology , Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
50
|
Kim MK, Shin H, Park KS, Kim H, Park J, Kim K, Nam J, Choo H, Chong Y. Benzimidazole Derivatives as Potent JAK1-Selective Inhibitors. J Med Chem 2015; 58:7596-602. [PMID: 26351728 DOI: 10.1021/acs.jmedchem.5b01263] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The Janus kinase (JAK) family comprises four members (JAK1, JAK2, JAK3, and Tyk2) that play a key role in mediating cytokine receptor signaling. JAK inhibition thus modulates cytokine-mediated effects. In particular, selective inhibition of JAK1 or JAK3 may provide an efficient therapeutic agent for the treatment of inflammatory diseases, with minimized side effects. In this study, as part of our continued efforts to develop a selective JAK1 inhibitor, a series of 1,2-disubstituted benzimidazole-5-carboxamide derivatives was prepared and their inhibitory activities against all four JAK isozymes were evaluated. A clear structure-activity relationship was observed with respect to JAK1 selectivity; this highlighted the importance of hydrogen bond donors at both N(1) and R2 positions located within a specific distance from the benzimidazole core. One of the synthesized compounds, 1-(2-aminoethyl)-2-(piperidin-4-yl)-1H-benzo[d]imidazole-5-carboxamide (5c), showed remarkable JAK1 selectivity (63-fold vs JAK2, 25-fold vs JAK3, and 74-fold vs Tyk2). Molecular docking revealed that the 2-aminoethyl and piperidin-4-yl substituents of 5c function as probes to differentiate the ATP-binding site of JAK1 from that of JAK2, resulting in preferential JAK1 binding. A kinase panel assay confirmed the JAK1 selectivity of 5c, which showed no appreciable inhibitory activity against 26 other protein kinases at 10 μM.
Collapse
Affiliation(s)
- Mi Kyoung Kim
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University , 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea
| | - Heerim Shin
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University , 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea
| | - Kwang-su Park
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University , 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea
| | - Hyungmi Kim
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University , 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea
| | - Jiseon Park
- R&D Center, Jeil Pharmaceutical Co., Ltd. 7 Cheongganggachangro, Baegam-myun, Cheoin-gu, Yongin-City, Kyunggi-Do 17172, Korea
| | - Kangjeon Kim
- R&D Center, Jeil Pharmaceutical Co., Ltd. 7 Cheongganggachangro, Baegam-myun, Cheoin-gu, Yongin-City, Kyunggi-Do 17172, Korea
| | - Joonwoo Nam
- R&D Center, Jeil Pharmaceutical Co., Ltd. 7 Cheongganggachangro, Baegam-myun, Cheoin-gu, Yongin-City, Kyunggi-Do 17172, Korea
| | - Hyunah Choo
- Center for Neuro-Medicine, Korea Institute of Science and Technology , 5, Hwarang-ro 14-gil, Seoungbuk-gu, Seoul 02792, Korea.,Department of Biological Chemistry, University of Science and Technology , 217 Gajeong-ro, Youseong-gu, Daejeon 34113, Korea
| | - Youhoon Chong
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University , 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea
| |
Collapse
|