1
|
Feng Y, Chen C, Shao A, Wu L, Hu H, Zhang T. Emerging interleukin-1 receptor-associated kinase 4 (IRAK4) inhibitors or degraders as therapeutic agents for autoimmune diseases and cancer. Acta Pharm Sin B 2024; 14:5091-5105. [PMID: 39807338 PMCID: PMC11725142 DOI: 10.1016/j.apsb.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/20/2024] [Accepted: 07/26/2024] [Indexed: 01/16/2025] Open
Abstract
Interleukin-1 receptor-related kinase (IRAK4) is a widely expressed serine/threonine kinase involved in the regulation of innate immunity. IRAK4 plays a pivotal role as a key kinase within the downstream signaling pathway cascades of interleukin-1 receptors (IL-1R) and Toll-like receptors (TLRs). The signaling pathways orchestrated by IRAK4 are integral to inflammatory responses, and its overexpression is implicated in the pathogenesis of inflammatory diseases, autoimmune disorders, and cancer. Consequently, targeting IRAK4-mediated signaling pathways has emerged as a promising therapeutic strategy. Small molecule inhibitors and degraders designed to modulate IRAK4 have shown efficacy in mitigating related diseases. In this paper, we will provide a detailed description of the structure and function of IRAK4, the role of IRAK4 in related diseases, as well as the currently reported small molecule inhibitors and degraders of IRAK4. It is expected to provide new directions for enriching the clinical treatment of inflammation and related diseases.
Collapse
Affiliation(s)
- Yifan Feng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Chengjuan Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Anqi Shao
- Department of Dermatology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lei Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Haiyu Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Tiantai Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
2
|
Zheng X, Ji N, Campbell V, Slavin A, Zhu X, Chen D, Rong H, Enerson B, Mayo M, Sharma K, Browne CM, Klaus CR, Li H, Massa G, McDonald AA, Shi Y, Sintchak M, Skouras S, Walther DM, Yuan K, Zhang Y, Kelleher J, Liu G, Luo X, Mainolfi N, Weiss MM. Discovery of KT-474─a Potent, Selective, and Orally Bioavailable IRAK4 Degrader for the Treatment of Autoimmune Diseases. J Med Chem 2024; 67:18022-18037. [PMID: 39151120 PMCID: PMC11513890 DOI: 10.1021/acs.jmedchem.4c01305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 08/18/2024]
Abstract
Interleukin-1 receptor associated kinase 4 (IRAK4) is an essential mediator of the IL-1R and TLR signaling pathways, both of which have been implicated in multiple autoimmune conditions. Hence, blocking the activity of IRAK4 represents an attractive approach for the treatment of autoimmune diseases. The activity of this serine/threonine kinase is dependent on its kinase and scaffolding activities; thus, degradation represents a potentially superior approach to inhibition. Herein, we detail the exploration of structure-activity relationships that ultimately led to the identification of KT-474, a potent, selective, and orally bioavailable heterobifunctional IRAK4 degrader. This represents the first heterobifunctional degrader evaluated in a nononcology indication and dosed to healthy human volunteers. This molecule successfully completed phase I studies in healthy adult volunteers and patients with atopic dermatitis or hidradenitis suppurativa. Phase II clinical trials in both of these indications have been initiated.
Collapse
Affiliation(s)
- Xiaozhang Zheng
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Nan Ji
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Veronica Campbell
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Anthony Slavin
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Xiao Zhu
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Dapeng Chen
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Haojing Rong
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Brad Enerson
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Michele Mayo
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Kirti Sharma
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Chris M. Browne
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Christine R. Klaus
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Haoran Li
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Ginny Massa
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Alice A. McDonald
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Yatao Shi
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Mike Sintchak
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Stephanie Skouras
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Dirk M. Walther
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Karen Yuan
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Yi Zhang
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Joe Kelleher
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Guang Liu
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Xinbo Luo
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Nello Mainolfi
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Matthew M. Weiss
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| |
Collapse
|
3
|
Yadav H, Shirumalla RK. Emerging trends in IRAK-4 kinase research. Mol Biol Rep 2023; 50:7825-7837. [PMID: 37490192 DOI: 10.1007/s11033-023-08438-w] [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: 05/27/2022] [Accepted: 04/06/2023] [Indexed: 07/26/2023]
Abstract
The IRAK-4 kinase lies at a critical signaling node that drives cancer cell survival through multiple mechanisms, activation, and translocation of NF-κB mediated inflammatory responses and innate immune signaling through regulation of interferon-α/β receptor (IFNα/β). Inhibition, of IRAK-4, has consequently drawn a lot of attention in recent years to address indications ranging from oncology to autoimmune disorders to neurodegeneration, etc. However, the key stumbling block in targeting IRAK-4 is that despite the inhibition of the kinase activity using an inhibitor the target remains effective, reducing the potential of an inhibitor. This is due to the "scaffolding effect" because of which although regulation of downstream processes by IRAK-4 has been primarily linked with kinase function; however, still, various reports have suggested that IRAK-4 has a non-kinase function in a variety of cell types. This is attributed to the myddosome complex formed by IRAK-4 with myd88, IRAK-2, and IRAK-1 which by itself can cause the activation of downstream effector TRAF6 despite inhibition of the kinase domain of IRAK-4. With this challenge, several groups initiated the development of targeting protein degraders of IRAK-4 using Proteolysis-Targeting Chimeras (PROTACs) technology to completely remove the IRAK-4 from the cellular milieu. In this review, we will capture all these developments and the evolving science around this target.
Collapse
Affiliation(s)
- Himanshu Yadav
- SGT College of Pharmacy, SGT University, Budhera, Gurugram, Haryana, 122505, India
| | - Raj Kumar Shirumalla
- SGT College of Pharmacy, SGT University, Budhera, Gurugram, Haryana, 122505, India.
| |
Collapse
|
4
|
Inami H, Mizutani T, Watanabe J, Hayashida H, Ito T, Terasawa T, Kontani T, Yamagishi H, Usuda H, Aoyama N, Imamura E, Ishikawa T. Design, synthesis, and pharmacological evaluation of N-(3-carbamoyl-1H-pyrazol-4-yl)-1,3-oxazole-4-carboxamide derivatives as interleukin-1 receptor-associated kinase 4 inhibitors with reduced potential for cytochrome P450 1A2 induction. Bioorg Med Chem 2023; 87:117302. [PMID: 37201454 DOI: 10.1016/j.bmc.2023.117302] [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: 03/09/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/20/2023]
Abstract
Interleukin-1 receptor-associated kinase 4 (IRAK4) is a critical molecule in Toll-like receptor/interleukin-1 receptor signaling and an attractive therapeutic target for a wide range of inflammatory and autoimmune diseases as well as cancers. In our search for novel IRAK4 inhibitors, we conducted structural modification of a thiazolecarboxamide derivative 1, a lead compound derived from high-throughput screening hits, to elucidate structure-activity relationship and improve drug metabolism and pharmacokinetic (DMPK) properties. First, conversion of the thiazole ring of 1 to an oxazole ring along with introduction of a methyl group at the 2-position of the pyridine ring aimed at reducing cytochrome P450 (CYP) inhibition were conducted to afford 16. Next, modification of the alkyl substituent at the 1-position of the pyrazole ring of 16 aimed at improving CYP1A2 induction properties revealed that branched alkyl and analogous substituents such as isobutyl (18) and (oxolan-3-yl)methyl (21), as well as six-membered saturated heterocyclic groups such as oxan-4-yl (2), piperidin-4-yl (24, 25), and dioxothian-4-y (26), are effective for reducing induction potential. Representative compound AS2444697 (2) exhibited potent IRAK4 inhibitory activity with an IC50 value of 20 nM and favorable DMPK properties such as low risk of drug-drug interactions mediated by CYPs as well as excellent metabolic stability and oral bioavailability.
Collapse
Affiliation(s)
- Hiroshi Inami
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan.
| | - Tsuyoshi Mizutani
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Junko Watanabe
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Hisashi Hayashida
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Tomonori Ito
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Takeshi Terasawa
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Toru Kontani
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Hiroaki Yamagishi
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Hiroyuki Usuda
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Naohiro Aoyama
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Emiko Imamura
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Takeshi Ishikawa
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| |
Collapse
|
5
|
Bhujbal SP, He W, Hah JM. Design of Novel IRAK4 Inhibitors Using Molecular Docking, Dynamics Simulation and 3D-QSAR Studies. Molecules 2022; 27:6307. [PMID: 36234844 PMCID: PMC9570937 DOI: 10.3390/molecules27196307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/13/2022] [Accepted: 09/21/2022] [Indexed: 02/05/2023] Open
Abstract
Treatment of several autoimmune diseases and types of cancer has been an intense area of research over the past two decades. Many signaling pathways that regulate innate and/or adaptive immunity, as well as those that induce overexpression or mutation of protein kinases, have been targeted for drug discovery. One of the serine/threonine kinases, Interleukin-1 Receptor Associated Kinase 4 (IRAK4) regulates signaling through various Toll-like receptors (TLRs) and interleukin-1 receptor (IL1R). It controls diverse cellular processes including inflammation, apoptosis, and cellular differentiation. MyD88 gain-of-function mutations or overexpression of IRAK4 has been implicated in various types of malignancies such as Waldenström macroglobulinemia, B cell lymphoma, colorectal cancer, pancreatic ductal adenocarcinoma, breast cancer, etc. Moreover, over activation of IRAK4 is also associated with several autoimmune diseases. The significant role of IRAK4 makes it an interesting target for the discovery and development of potent small molecule inhibitors. A few potent IRAK4 inhibitors such as PF-06650833, RA9 and BAY1834845 have recently entered phase I/II clinical trial studies. Nevertheless, there is still a need of selective inhibitors for the treatment of cancer and various autoimmune diseases. A great need for the same intrigued us to perform molecular modeling studies on 4,6-diaminonicotinamide derivatives as IRAK4 inhibitors. We performed molecular docking and dynamics simulation of 50 ns for one of the most active compounds of the dataset. We also carried out MM-PBSA binding free energy calculation to identify the active site residues, interactions of which are contributing to the total binding energy. The final 50 ns conformation of the most active compound was selected to perform dataset alignment in a 3D-QSAR study. Generated RF-CoMFA (q2 = 0.751, ONC = 4, r2 = 0.911) model revealed reasonable statistical results. Overall results of molecular dynamics simulation, MM-PBSA binding free energy calculation and RF-CoMFA model revealed important active site residues of IRAK4 and necessary structural properties of ligand to design more potent IRAK4 inhibitors. We designed few IRAK4 inhibitors based on these results, which possessed higher activity (predicted pIC50) than the most active compounds of the dataset selected for this study. Moreover, ADMET properties of these inhibitors revealed promising results and need to be validated using experimental studies.
Collapse
Affiliation(s)
- Swapnil P. Bhujbal
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 426-791, Korea
- Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 426-791, Korea
| | - Weijie He
- Second Affiliated Hospital of Shantou University Medical College, Shantou University, Shantou 515000, China
| | - Jung-Mi Hah
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 426-791, Korea
- Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 426-791, Korea
| |
Collapse
|
6
|
Chen Y, Tso K, Heckrodt TJ, Li H, Yen R, Lin N, Singh R, Taylor V, Masuda ES, Park G, Payan DG. Bicyclic pyrimidine compounds as potent IRAK4 inhibitors. Bioorg Med Chem Lett 2022; 73:128900. [PMID: 35863718 DOI: 10.1016/j.bmcl.2022.128900] [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: 05/03/2022] [Revised: 07/07/2022] [Accepted: 07/14/2022] [Indexed: 12/01/2022]
Abstract
Interleukin-1 receptor-associated kinase 4 (IRAK4) plays a critical role in transduction of IL-1R/TLR signaling which is responsible for innate immune response. From HTS campaign, bicyclic-pyrimidine compounds have been identified as potent IRAK4 inhibitors, exhibiting good potency in both IRAK4 biochemical and LPS induced IL-23 inhibition cell-based assays. The SAR efforts were focused on further improving on-target potency, reducing PAD activities of HTS hit molecule and improving in vivo PK profiles of early lead compounds. When different aromatic rings were fused to the pyrimidine core, and with various substituents at 2- or 4-position of the pyrimidine, the impact on potency and PK properties were observed and are discussed. Selected compounds were further evaluated in IL-1β induced IL-6 inhibition acute animal model and rodent arthritis disease model, of which compounds 33 and 39 showed good efficacy in both studies.
Collapse
Affiliation(s)
- Yan Chen
- Rigel Pharmaceuticals Inc, 1180 Veterans Boulevard, South San Francisco, CA 94080, United States.
| | - Kin Tso
- Rigel Pharmaceuticals Inc, 1180 Veterans Boulevard, South San Francisco, CA 94080, United States
| | - Thilo J Heckrodt
- Rigel Pharmaceuticals Inc, 1180 Veterans Boulevard, South San Francisco, CA 94080, United States
| | - Hui Li
- Rigel Pharmaceuticals Inc, 1180 Veterans Boulevard, South San Francisco, CA 94080, United States
| | - Rose Yen
- Rigel Pharmaceuticals Inc, 1180 Veterans Boulevard, South San Francisco, CA 94080, United States
| | - Nan Lin
- Rigel Pharmaceuticals Inc, 1180 Veterans Boulevard, South San Francisco, CA 94080, United States
| | - Rajinder Singh
- Rigel Pharmaceuticals Inc, 1180 Veterans Boulevard, South San Francisco, CA 94080, United States
| | - Vanessa Taylor
- Rigel Pharmaceuticals Inc, 1180 Veterans Boulevard, South San Francisco, CA 94080, United States
| | - Esteban S Masuda
- Rigel Pharmaceuticals Inc, 1180 Veterans Boulevard, South San Francisco, CA 94080, United States
| | - Gary Park
- Rigel Pharmaceuticals Inc, 1180 Veterans Boulevard, South San Francisco, CA 94080, United States
| | - Donald G Payan
- Rigel Pharmaceuticals Inc, 1180 Veterans Boulevard, South San Francisco, CA 94080, United States
| |
Collapse
|
7
|
Cumming IA, Degorce SL, Aagaard A, Braybrooke EL, Davies NL, Diène CR, Eatherton AJ, Felstead HR, Groombridge SD, Lenz EM, Li Y, Nai Y, Pearson S, Robb GR, Scott JS, Steward OR, Wu C, Xue Y, Zhang L, Zhang Y. Identification and optimisation of a pyrimidopyridone series of IRAK4 inhibitors. Bioorg Med Chem 2022; 63:116729. [PMID: 35439688 DOI: 10.1016/j.bmc.2022.116729] [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: 01/13/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 12/01/2022]
Abstract
In this article, we report the discovery of a series of pyrimidopyridones as inhibitors of IRAK4 kinase. From a previously disclosed 5-azaquinazoline series, we found that switching the pyridine ring for an N-substituted pyridone gave a novel hinge binding scaffold which retained potency against IRAK4. Importantly, introduction of the carbonyl established an internal hydrogen bond with the 4-NH, establishing a conformational lock and allowing truncation of the large basic substituent to a 1-methylcyclopyl group. Subsequent optimisation, facilitated by X-ray crystal structures, allowed identification of preferred substituents at both the pyridone core and pyrazole. Subsequent combinations of optimal groups allowed control of lipophilicity and identification of potent and selective inhibitors of IRAK4 with better in vitro permeability and lower clearance.
Collapse
Affiliation(s)
- Iain A Cumming
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom.
| | - Sébastien L Degorce
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Anna Aagaard
- Discovery Sciences, R&D, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| | - Erin L Braybrooke
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Nichola L Davies
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Coura R Diène
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Andrew J Eatherton
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Hannah R Felstead
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Sam D Groombridge
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Eva M Lenz
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Yunxia Li
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176 PR China
| | - Youfeng Nai
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176 PR China
| | - Stuart Pearson
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Graeme R Robb
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - James S Scott
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Oliver R Steward
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Chengyan Wu
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176 PR China
| | - Yafeng Xue
- Discovery Sciences, R&D, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| | - Lanping Zhang
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176 PR China
| | - Yanxiu Zhang
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176 PR China
| |
Collapse
|
8
|
Chen Y, Singh R, Lin N, Taylor V, Masuda ES, Payan DG. Discovery of 5-Aryl-2,4-diaminopyrimidine Compounds as Potent and Selective IRAK4 Inhibitors. ACS Med Chem Lett 2022; 13:714-719. [PMID: 35450353 PMCID: PMC9014513 DOI: 10.1021/acsmedchemlett.2c00056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/29/2022] [Indexed: 12/17/2022] Open
Abstract
IRAK4 kinase plays a key role in TLR/IL-1R signaling pathways that regulate innate immune responses, and if uncontrolled, it is responsible for various inflammatory disorders. By high-throughput screening (HTS) and hit-to-lead optimization, compounds with a 5-aryl-2,4-diaminopyrimidine core structure have been identified as potent IRAK4 inhibitors. A cocrystal structure of IRAK4 protein with an early lead molecule helped with understanding the structure-activity relationship and the design of the new compounds. Initial HTS hits from this series of compounds were also found to inhibit TAK1 kinase, which would cause liver toxicity and potentially bone marrow failure. Optimization of this series resulted in improved selectivity over TAK1 kinase. The TAK1 selectivity was found to be closely associated with different sizes and types of substituents at the 5-position of the pyrimidine. The impact of other pyrimidine substituents on the potency and selectivity was also explored. A few representative compounds were evaluated in IL-1β-induced IL-6 inhibition animal model studies and showed modest efficacy.
Collapse
Affiliation(s)
- Yan Chen
- Rigel Pharmaceuticals Inc., 1180 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Rajinder Singh
- Rigel Pharmaceuticals Inc., 1180 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Nan Lin
- Rigel Pharmaceuticals Inc., 1180 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Vanessa Taylor
- Rigel Pharmaceuticals Inc., 1180 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Esteban S. Masuda
- Rigel Pharmaceuticals Inc., 1180 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Donald G. Payan
- Rigel Pharmaceuticals Inc., 1180 Veterans Boulevard, South San Francisco, California 94080, United States
| |
Collapse
|
9
|
Singh S, Utreja D, Kumar V. Pyrrolo[2,1-f][1,2,4]triazine: a promising fused heterocycle to target kinases in cancer therapy. Med Chem Res 2021; 31:1-25. [PMID: 34803342 PMCID: PMC8590428 DOI: 10.1007/s00044-021-02819-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 10/29/2021] [Indexed: 12/21/2022]
Abstract
Cancer is the second leading cause of death worldwide responsible for about 10 million deaths per year. To date several approaches have been developed to treat this deadly disease including surgery, chemotherapy, radiation therapy, hormonal therapy, targeted therapy, and synthetic lethality. The targeted therapy refers to targeting only specific proteins or enzymes that are dysregulated in cancer rather than killing all rapidly dividing cells, has gained much attention in the recent past. Kinase inhibition is one of the most successful approaches in targeted therapy. As of 30 March 2021, FDA has approved 65 small molecule protein kinase inhibitors and most of them are for cancer therapy. Interestingly, several kinase inhibitors contain one or more fused heterocycles as part of their structures. Pyrrolo[2,1-f][1,2,4]triazine is one the most interesting fused heterocycle that is an integral part of several kinase inhibitors and nucleoside drugs viz. avapritinib and remdesivir. This review articles focus on the recent advances made in the development of kinase inhibitors containing pyrrolo[2,1-f][1,2,4]triazine scaffold. ![]()
Collapse
Affiliation(s)
- Sarbjit Singh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Divya Utreja
- Department of Chemistry, Punjab Agricultural University, Ludhiana, 141004 Punjab India
| | - Vimal Kumar
- Department of Chemistry, Dr B. R. Ambedkar National Institute of Technology (NIT), Jalandhar, 144011 Punjab India
| |
Collapse
|
10
|
Umar S, Palasiewicz K, Van Raemdonck K, Volin MV, Romay B, Amin MA, Zomorrodi RK, Arami S, Gonzalez M, Rao V, Zanotti B, Fox DA, Sweiss N, Shahrara S. IRAK4 inhibition: a promising strategy for treating RA joint inflammation and bone erosion. Cell Mol Immunol 2021; 18:2199-2210. [PMID: 32415262 PMCID: PMC8429735 DOI: 10.1038/s41423-020-0433-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/30/2020] [Indexed: 01/22/2023] Open
Abstract
Flares of joint inflammation and resistance to currently available biologic therapeutics in rheumatoid arthritis (RA) patients could reflect activation of innate immune mechanisms. Herein, we show that a TLR7 GU-rich endogenous ligand, miR-Let7b, potentiates synovitis by amplifying RA monocyte and fibroblast (FLS) trafficking. miR-Let7b ligation to TLR7 in macrophages (MΦs) and FLSs expanded the synovial inflammatory response. Moreover, secretion of M1 monokines triggered by miR-Let7b enhanced Th1/Th17 cell differentiation. We showed that IRAK4 inhibitor (i) therapy attenuated RA disease activity by blocking TLR7-induced M1 MΦ or FLS activation, as well as monokine-modulated Th1/Th17 cell polarization. IRAK4i therapy also disrupted RA osteoclastogenesis, which was amplified by miR-Let7b ligation to joint myeloid TLR7. Hence, the effectiveness of IRAK4i was compared with that of a TNF inhibitor (i) or anti-IL-6R treatment in collagen-induced arthritis (CIA) and miR-Let7b-mediated arthritis. We found that TNF or IL-6R blocking therapies mitigated CIA by reducing the infiltration of joint F480+iNOS+ MΦs, the expression of certain monokines, and Th1 cell differentiation. Unexpectedly, these biologic therapies were unable to alleviate miR-Let7b-induced arthritis. The superior efficacy of IRAK4i over anti-TNF or anti-IL-6R therapy in miR-Let7b-induced arthritis or CIA was due to the ability of IRAK4i therapy to restrain the migration of joint F480+iNOS+ MΦs, vimentin+ fibroblasts, and CD3+ T cells, in addition to negating the expression of a wide range of monokines, including IL-12, MIP2, and IRF5 and Th1/Th17 lymphokines. In conclusion, IRAK4i therapy may provide a promising strategy for RA therapy by disconnecting critical links between inflammatory joint cells.
Collapse
Affiliation(s)
- Sadiq Umar
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Karol Palasiewicz
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Katrien Van Raemdonck
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Michael V Volin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA
| | - Bianca Romay
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - M Asif Amin
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI, 481096, USA
| | - Ryan K Zomorrodi
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Shiva Arami
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mark Gonzalez
- Department of Orthopedic Surgery, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Vikram Rao
- Pfizer Research, Cambridge, MA, 02139, USA
| | - Brian Zanotti
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA
| | - David A Fox
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI, 481096, USA
| | - Nadera Sweiss
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA.
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
11
|
Degorce SL, Aagaard A, Anjum R, Cumming IA, Diène CR, Fallan C, Johnson T, Leuchowius KJ, Orton AL, Pearson S, Robb GR, Rosen A, Scarfe GB, Scott JS, Smith JM, Steward OR, Terstiege I, Tucker MJ, Turner P, Wilkinson SD, Wrigley GL, Xue Y. Improving metabolic stability and removing aldehyde oxidase liability in a 5-azaquinazoline series of IRAK4 inhibitors. Bioorg Med Chem 2020; 28:115815. [PMID: 33091850 DOI: 10.1016/j.bmc.2020.115815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/07/2020] [Indexed: 11/29/2022]
Abstract
In this article, we report our efforts towards improving in vitro human clearance in a series of 5-azaquinazolines through a series of C4 truncations and C2 expansions. Extensive DMPK studies enabled us to tackle high Aldehyde Oxidase (AO) metabolism and unexpected discrepancies in human hepatocyte and liver microsomal intrinsic clearance. Our efforts culminated with the discovery of 5-azaquinazoline 35, which also displayed exquisite selectivity for IRAK4, and showed synergistic in vitro activity against MyD88/CD79 double mutant ABC-DLBCL in combination with the covalent BTK inhibitor acalabrutinib.
Collapse
Affiliation(s)
- Sébastien L Degorce
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Boston, 35 Gatehouse Drive, Waltham, MA 02451, United States.
| | - Anna Aagaard
- Discovery Sciences, R&D, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| | - Rana Anjum
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Boston, 35 Gatehouse Drive, Waltham, MA 02451, United States
| | - Iain A Cumming
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Coura R Diène
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Charlene Fallan
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Tony Johnson
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | | | - Alexandra L Orton
- DMPK, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Stuart Pearson
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Graeme R Robb
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Alan Rosen
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Boston, 35 Gatehouse Drive, Waltham, MA 02451, United States
| | - Graeme B Scarfe
- DMPK, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - James S Scott
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - James M Smith
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Oliver R Steward
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Ina Terstiege
- Medicinal Chemistry, R&I, R&D, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| | - Michael J Tucker
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Paul Turner
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Stephen D Wilkinson
- DMPK, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Gail L Wrigley
- Medicinal Chemistry, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, United Kingdom
| | - Yafeng Xue
- Discovery Sciences, R&D, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| |
Collapse
|
12
|
Hatcher JM, Yang G, Wang L, Ficarro SB, Buhrlage S, Wu H, Marto JA, Treon SP, Gray NS. Discovery of a Selective, Covalent IRAK1 Inhibitor with Antiproliferative Activity in MYD88 Mutated B-Cell Lymphoma. ACS Med Chem Lett 2020; 11:2238-2243. [PMID: 33214835 DOI: 10.1021/acsmedchemlett.0c00378] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/06/2020] [Indexed: 12/30/2022] Open
Abstract
Interleukin 1 (IL-1) receptor-associated kinases (IRAKs) are serine/threonine kinases that play critical roles in initiating the innate immune response against foreign pathogens. Additionally, dysregulation of IRAK1 signaling plays a role in neoplastic disorders. For example, IRAK1 was shown to be important for survival and proliferation in many B-cell lymphomas, including Waldenström's macroglobulinemia (WM) and ABC subtype Diffused Large B-cell Lymphoma (DLBCL) cells. Here, we report the discovery of a highly potent and selective covalent inhibitor of IRAK1, JH-X-119-01. Intact protein MS labeling studies confirmed that JH-X-119-01 irreversibly labels IRAK1 at C302. This compound exhibited cytotoxic activity at single digit micromolar concentrations in a panel of WM, DLBCL, and lymphoma cell lines expressing MYD88. Cotreatment of JH-X-119-01 with the BTK inhibitor ibrutinib resulted in synergistic killing effects in these systems. Taken together, JH-X-119-01 represents a highly selective probe of IRAK1 for further development.
Collapse
Affiliation(s)
- John M. Hatcher
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, United States
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, 360 Longwood Ave, Longwood Center LC-2209, Boston, Massachusetts 02115, United States
| | - Guang Yang
- Bing Center for Waldenstrom’s Macroglobulinemia, Dana Farber Cancer Institute, Boston, Massachusetts 02115, United States
- Department of Medical Oncology, Dana Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Li Wang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
| | - Scott B. Ficarro
- Department of Cancer Biology, Department of Oncologic Pathology, Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, United States
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Sara Buhrlage
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, United States
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, 360 Longwood Ave, Longwood Center LC-2209, Boston, Massachusetts 02115, United States
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
| | - Jarrod A. Marto
- Department of Cancer Biology, Department of Oncologic Pathology, Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, United States
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Steven P. Treon
- Bing Center for Waldenstrom’s Macroglobulinemia, Dana Farber Cancer Institute, Boston, Massachusetts 02115, United States
- Department of Medical Oncology, Dana Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Nathanael S. Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, United States
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, 360 Longwood Ave, Longwood Center LC-2209, Boston, Massachusetts 02115, United States
| |
Collapse
|
13
|
Rajapaksa NS, Gobbi A, Drobnick J, Do S, Kolesnikov A, Liang J, Chen Y, Sujatha-Bhaskar S, Huang Z, Brightbill H, Francis R, Yu C, Choo EF, DeMent K, Ran Y, An L, Emson C, Maher J, Wai J, McKenzie BS, Lupardus PJ, Zarrin AA, Kiefer JR, Bryan MC. Discovery of Potent Benzolactam IRAK4 Inhibitors with Robust in Vivo Activity. ACS Med Chem Lett 2020; 11:327-333. [PMID: 32184965 DOI: 10.1021/acsmedchemlett.9b00380] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/11/2019] [Indexed: 12/17/2022] Open
Abstract
IRAK4 kinase activity transduces signaling from multiple IL-1Rs and TLRs to regulate cytokines and chemokines implicated in inflammatory diseases. As such, there is high interest in identifying selective IRAK4 inhibitors for the treatment of these disorders. We previously reported the discovery of potent and selective dihydrobenzofuran inhibitors of IRAK4. Subsequent studies, however, showed inconsistent inhibition in disease-relevant pharmacodynamic models. Herein, we describe application of a human whole blood assay to the discovery of a series of benzolactam IRAK4 inhibitors. We identified potent molecule 19 that achieves robust in vivo inhibition of cytokines relevant to human disease.
Collapse
Affiliation(s)
- Naomi S. Rajapaksa
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Alberto Gobbi
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Joy Drobnick
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Steven Do
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Aleksandr Kolesnikov
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Jun Liang
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Yongsheng Chen
- WuXi Apptech, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, P. R. China
| | | | - Zhiyu Huang
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Hans Brightbill
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Ross Francis
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Christine Yu
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Edna F. Choo
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Kevin DeMent
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Yingqing Ran
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Le An
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Claire Emson
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Jonathan Maher
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - John Wai
- WuXi Apptech, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, P. R. China
| | - Brent S. McKenzie
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Patrick J. Lupardus
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Ali A. Zarrin
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - James R. Kiefer
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| | - Marian C. Bryan
- Genentech, Inc., One DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
14
|
Degorce SL, Anjum R, Bloecher A, Carbajo RJ, Dillman KS, Drew L, Halsall CT, Lenz EM, Lindsay NA, Mayo MF, Pink JH, Robb GR, Rosen A, Scott JS, Xue Y. Discovery of a Series of 5-Azaquinazolines as Orally Efficacious IRAK4 Inhibitors Targeting MyD88L265P Mutant Diffuse Large B Cell Lymphoma. J Med Chem 2019; 62:9918-9930. [DOI: 10.1021/acs.jmedchem.9b01346] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Sébastien L. Degorce
- Medicinal Chemistry, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, U.K
| | - Rana Anjum
- Bioscience, Oncology R&D, AstraZeneca, Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Andrew Bloecher
- Bioscience, Oncology R&D, AstraZeneca, Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Rodrigo J. Carbajo
- Medicinal Chemistry, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, U.K
| | - Keith S. Dillman
- Bioscience, Oncology R&D, AstraZeneca, Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Lisa Drew
- Bioscience, Oncology R&D, AstraZeneca, Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Christopher T. Halsall
- Medicinal Chemistry, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, U.K
| | - Eva M. Lenz
- Medicinal Chemistry, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, U.K
| | - Nicola A. Lindsay
- Medicinal Chemistry, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, U.K
| | - Michele F. Mayo
- Bioscience, Oncology R&D, AstraZeneca, Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Jennifer H. Pink
- Medicinal Chemistry, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, U.K
| | - Graeme R. Robb
- Medicinal Chemistry, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, U.K
| | - Alan Rosen
- Bioscience, Oncology R&D, AstraZeneca, Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - James S. Scott
- Medicinal Chemistry, Oncology R&D, AstraZeneca, Cambridge Science Park, Unit 310 Darwin Building, Cambridge CB4 0WG, U.K
| | - Yafeng Xue
- Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-431 83, Mölndal, Sweden
| |
Collapse
|
15
|
Targeting IRAK4 disrupts inflammatory pathways and delays tumor development in chronic lymphocytic leukemia. Leukemia 2019; 34:100-114. [PMID: 31197259 PMCID: PMC8075947 DOI: 10.1038/s41375-019-0507-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/04/2019] [Accepted: 04/26/2019] [Indexed: 12/20/2022]
Abstract
Interleukin-1 receptor-associated kinase 4 (IRAK4) plays a critical role in Toll-like receptor (TLR) signal transduction and innate immune responses. Recruitment and subsequent activation of IRAK4 upon TLR stimulation is mediated by the myeloid differentiation primary response 88 (MYD88) adaptor protein. Around 3% of chronic lymphocytic leukemia (CLL) patients have activating mutations of MYD88, a driver mutation in this disease. Here, we studied the effects of TLR activation and the pharmacological inhibition of IRAK4 with ND2158, an IRAK4 competitive inhibitor, as a therapeutic approach in CLL. Our in vitro studies demonstrated that ND2158 preferentially killed CLL cells in a dose-dependent manner. We further observed a decrease in NF-κB and STAT3 signaling, cytokine secretion, proliferation and migration of primary CLL cells from MYD88-mutated and -unmutated cases. In the Eµ-TCL1 adoptive transfer mouse model of CLL, ND2158 delayed tumor progression and modulated the activity of myeloid and T cells. Our findings show the importance of TLR signaling in CLL development and suggest IRAK4 as a therapeutic target for this disease.
Collapse
|
16
|
McElroy WT. Interleukin-1 receptor-associated kinase 4 (IRAK4) inhibitors: an updated patent review (2016-2018). Expert Opin Ther Pat 2019; 29:243-259. [PMID: 30916602 DOI: 10.1080/13543776.2019.1597850] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Interleukin-1 receptor-associated kinase 4 (IRAK4) is the most upstream kinase in Toll/Interleukin-1 receptor (TIR) signaling. Human and rodent genetics support the role of IRAK4 in immune function and the involvement of IRAK4-dependent signaling in certain cancers is hypothesized. The accumulating evidence has motivated the discovery of IRAK4 inhibitors that could be used therapeutically. AREAS COVERED This review summarizes patents published in 2016-2018 claiming IRAK4 inhibitors. Representative analogues from each patent are presented with a focus on compounds that have been profiled in cellular and in vivo assays. EXPERT OPINION The last three years have seen an increased number of IRAK4 inhibitors with which to assess the therapeutic potential of the target. At least 5 companies are believed to have advanced to the clinic. Pfizer is in phase II for rheumatoid arthritis (RA). The outcomes of these studies should inform on the therapeutic potential in autoimmune disease and cancer.
Collapse
Affiliation(s)
- William T McElroy
- a Merck Sharp & Dohme Corp , a subsidiary of Merck & Co., Inc , Kenilworth , NJ , USA
| |
Collapse
|
17
|
Singer JW, Fleischman A, Al-Fayoumi S, Mascarenhas JO, Yu Q, Agarwal A. Inhibition of interleukin-1 receptor-associated kinase 1 (IRAK1) as a therapeutic strategy. Oncotarget 2018; 9:33416-33439. [PMID: 30279971 PMCID: PMC6161786 DOI: 10.18632/oncotarget.26058] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 08/15/2018] [Indexed: 02/06/2023] Open
Abstract
Interleukin-1 receptor-associated kinases (IRAK1, IRAK2, IRAK3 [IRAK-M], and IRAK4) are serine-threonine kinases involved in toll-like receptor and interleukin-1 signaling pathways, through which they regulate innate immunity and inflammation. Evidence exists that IRAKs play key roles in the pathophysiologies of cancers, and metabolic and inflammatory diseases, and that IRAK inhibition has potential therapeutic benefits. Molecules capable of selectively interfering with IRAK function and expression have been reported, paving the way for the clinical evaluation of IRAK inhibition. Herein, we focus on IRAK1, review its structure and physiological roles, and summarize emerging data for IRAK1 inhibitors in preclinical and clinical studies.
Collapse
Affiliation(s)
| | - Angela Fleischman
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | | | - John O. Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qiang Yu
- Genome Institute of Singapore, Singapore, SG, Singapore
| | - Anupriya Agarwal
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
18
|
Elkon KB. Review: Cell Death, Nucleic Acids, and Immunity: Inflammation Beyond the Grave. Arthritis Rheumatol 2018; 70:805-816. [PMID: 29439290 PMCID: PMC5984680 DOI: 10.1002/art.40452] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 02/08/2018] [Indexed: 12/29/2022]
Abstract
Cells of the innate immune system are rigged with sensors that detect nucleic acids derived from microbes, especially viruses. It has become clear that these same sensors that respond to nucleic acids derived from damaged cells or defective intracellular processing are implicated in triggering diseases such as lupus and arthritis. The ways in which cells die and the concomitant presence of proteins and peptides that allow nucleic acids to re-enter cells profoundly influence innate immune responses. In this review, we briefly discusses different types of programmed necrosis, such as pyroptosis, necroptosis, and NETosis, and explains how nucleic acids can engage intracellular receptors and stimulate inflammation. Host protective mechanisms that include compartmentalization of receptors and nucleases as well as the consequences of nuclease deficiencies are explored. In addition, proximal and distal targets in the nucleic acid stimulation of inflammation are discussed in terms of their potential amenability to therapy for the attenuation of innate immune activation and disease pathogenesis.
Collapse
Affiliation(s)
- Keith B. Elkon
- Department of Medicine and Immunology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
19
|
Degorce SL, Anjum R, Dillman KS, Drew L, Groombridge SD, Halsall CT, Lenz EM, Lindsay NA, Mayo MF, Pink JH, Robb GR, Scott JS, Stokes S, Xue Y. Optimization of permeability in a series of pyrrolotriazine inhibitors of IRAK4. Bioorg Med Chem 2018; 26:913-924. [DOI: 10.1016/j.bmc.2018.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/10/2018] [Accepted: 01/12/2018] [Indexed: 10/18/2022]
|
20
|
Scott JS, Degorce SL, Anjum R, Culshaw J, Davies RDM, Davies NL, Dillman KS, Dowling JE, Drew L, Ferguson AD, Groombridge SD, Halsall CT, Hudson JA, Lamont S, Lindsay NA, Marden SK, Mayo MF, Pease JE, Perkins DR, Pink JH, Robb GR, Rosen A, Shen M, McWhirter C, Wu D. Discovery and Optimization of Pyrrolopyrimidine Inhibitors of Interleukin-1 Receptor Associated Kinase 4 (IRAK4) for the Treatment of Mutant MYD88L265P Diffuse Large B-Cell Lymphoma. J Med Chem 2017; 60:10071-10091. [DOI: 10.1021/acs.jmedchem.7b01290] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- James S. Scott
- Oncology,
IMED Biotech Unit, AstraZeneca, Cambridge CB4 0FZ, United Kingdom
| | | | - Rana Anjum
- Oncology,
IMED Biotech Unit, AstraZeneca, Boston, Massachusetts 02451, United States
| | - Janet Culshaw
- Oncology,
IMED Biotech Unit, AstraZeneca, Macclesfield SK10 4TG, United Kingdom
| | - Robert D. M. Davies
- Oncology,
IMED Biotech Unit, AstraZeneca, Macclesfield SK10 4TG, United Kingdom
| | - Nichola L. Davies
- Oncology,
IMED Biotech Unit, AstraZeneca, Cambridge CB4 0FZ, United Kingdom
| | - Keith S. Dillman
- Oncology,
IMED Biotech Unit, AstraZeneca, Boston, Massachusetts 02451, United States
| | - James E. Dowling
- Oncology,
IMED Biotech Unit, AstraZeneca, Boston, Massachusetts 02451, United States
| | - Lisa Drew
- Oncology,
IMED Biotech Unit, AstraZeneca, Boston, Massachusetts 02451, United States
| | - Andrew D. Ferguson
- Oncology,
IMED Biotech Unit, AstraZeneca, Boston, Massachusetts 02451, United States
| | - Sam D. Groombridge
- Oncology,
IMED Biotech Unit, AstraZeneca, Macclesfield SK10 4TG, United Kingdom
| | | | - Julian A. Hudson
- Oncology,
IMED Biotech Unit, AstraZeneca, Macclesfield SK10 4TG, United Kingdom
| | - Scott Lamont
- Oncology,
IMED Biotech Unit, AstraZeneca, Cambridge CB4 0FZ, United Kingdom
| | - Nicola A. Lindsay
- Oncology,
IMED Biotech Unit, AstraZeneca, Cambridge CB4 0FZ, United Kingdom
| | - Stacey K. Marden
- Pharmaceutical
Sciences, IMED Biotech Unit, AstraZeneca, Boston, Massachusetts 02451, United States
| | - Michele F. Mayo
- Oncology,
IMED Biotech Unit, AstraZeneca, Boston, Massachusetts 02451, United States
| | - J. Elizabeth Pease
- Oncology,
IMED Biotech Unit, AstraZeneca, Cambridge CB4 0FZ, United Kingdom
| | - David R. Perkins
- Oncology,
IMED Biotech Unit, AstraZeneca, Macclesfield SK10 4TG, United Kingdom
| | - Jennifer H. Pink
- Oncology,
IMED Biotech Unit, AstraZeneca, Macclesfield SK10 4TG, United Kingdom
| | - Graeme R. Robb
- Oncology,
IMED Biotech Unit, AstraZeneca, Cambridge CB4 0FZ, United Kingdom
| | - Alan Rosen
- Oncology,
IMED Biotech Unit, AstraZeneca, Boston, Massachusetts 02451, United States
| | - Minhui Shen
- Oncology,
IMED Biotech Unit, AstraZeneca, Boston, Massachusetts 02451, United States
| | - Claire McWhirter
- Oncology,
IMED Biotech Unit, AstraZeneca, Cambridge CB4 0FZ, United Kingdom
| | - Dedong Wu
- Pharmaceutical
Sciences, IMED Biotech Unit, AstraZeneca, Boston, Massachusetts 02451, United States
| |
Collapse
|