1
|
Feng D, Liu B, Chen Z, Xu J, Geng M, Duan W, Ai J, Zhang H. Discovery of hematopoietic progenitor kinase 1 inhibitors using machine learning-based screening and free energy perturbation. J Biomol Struct Dyn 2025; 43:4152-4164. [PMID: 38198294 DOI: 10.1080/07391102.2024.2301754] [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: 09/12/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024]
Abstract
Hematopoietic progenitor kinase 1 (HPK1) is a key negative regulator of T-cell receptor (TCR) signaling and a promising target for cancer immunotherapy. The development of novel HPK1 inhibitors is challenging yet promising. In this study, we used a combination of machine learning (ML)-based virtual screening and free energy perturbation (FEP) calculations to identify novel HPK1 inhibitors. ML-based screening yielded 10 potent HPK1 inhibitors (IC50 < 1 μM). The FEP-guided modification of the in-house false-positive hit, DW21302, revealed that a single key atom change could trigger activity cliffs. The resulting DW21302-A was a potent HPK1 inhibitor (IC50 = 2.1 nM) and potently inhibited cellular HPK1 signaling and enhanced T-cell function. Molecular dynamics (MD) simulations and ADME predictions confirmed DW21302-A as candidate compound. This study provides new strategies and chemical scaffolds for HPK1 inhibitor development.
Collapse
Affiliation(s)
- Dazhi Feng
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Bo Liu
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| | - Zhiwei Chen
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Meiyu Geng
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Wenhu Duan
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Jing Ai
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hefeng Zhang
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
2
|
Zhang T, Sun Y, Xia J, Fan H, Shi D, Wu Q, Huang M, Hou XY. Targeting HPK1 inhibits neutrophil responses to mitigate post-stroke lung and cerebral injuries. EMBO Mol Med 2025:10.1038/s44321-025-00220-8. [PMID: 40169896 DOI: 10.1038/s44321-025-00220-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 03/01/2025] [Accepted: 03/10/2025] [Indexed: 04/03/2025] Open
Abstract
Circulating neutrophils are responsible for poor neurological outcomes and have been implicated in respiratory morbidity after acute ischemic stroke (AIS). However, the molecular mechanisms regulating neutrophil responses and their pathological relevance in post-stroke complications remain unclear. In this study, we investigated the involvement of hematopoietic progenitor kinase 1 (HPK1) in neutrophil responses and mobilization, as well as subsequent lung and cerebral injuries following AIS. We found that lipopolysaccharide treatment triggered neutrophil activation in an HPK1-dependent manner. HPK1 enhanced intrinsic NF-κB/STAT3/p38-MAPK pathways and gasdermin D cleavage, leading to neutrophil hyperactivation. Following AIS, HPK1 promoted the mobilization of CXCR2high bone marrow neutrophils. HPK1 loss inhibited peripheral neutrophil hyperactivation, neutrophil infiltration, and aggregation of neutrophil extracellular traps, progressively alleviating systemic inflammation and impairments in mouse pulmonary and neurological functions. Furthermore, HPK1 pharmacological inhibition attenuated post-stroke pulmonary and neurological impairments in mice. Our findings revealed that HPK1 upregulates neutrophil mobilization and various responses, promoting post-stroke systemic inflammation and tissue injury. This study highlights HPK1 as a therapeutic target for improving pulmonary and neurological functions after AIS.
Collapse
Affiliation(s)
- Tingting Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Ying Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Jing Xia
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Hongye Fan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Dingfang Shi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Qian Wu
- The Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Ming Huang
- Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| | - Xiao-Yu Hou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
3
|
Yuan YH, Mao JY, Yue JF, He ML, Hui Z, Yin H, Wang J, Ye XY. Updated patent review for hematopoietic progenitor kinase (HPK1) inhibitors and degraders (2021-present). Expert Opin Ther Pat 2025; 35:387-408. [PMID: 39950624 DOI: 10.1080/13543776.2025.2462834] [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: 05/06/2024] [Accepted: 01/29/2025] [Indexed: 02/18/2025]
Abstract
INTRODUCTION Hematopoietic progenitor cell kinase (HPK1) is a serine/threonine kinase of MAP4K family. It negatively regulates T cell receptor and B cell signal transduction. The loss of HPK1 kinase function increases the secretion of cytokines and enhances T cell signal transduction, virus clearance and tumor growth inhibition. Therefore, HPK1 is considered as a promising drug target for tumor immunotherapy. AREA COVERED This article surveys the patents published since 2021 aiming to analyze the structural features of scaffolds and the patent landscape. It also discusses the recent clinical developments and provides perspectives on the challenges and the future directions. EXPERT OPINION HPK1 kinase is a viable drug target, and there is an increasing number of clinical studies on HPK1 inhibitors. In the clinical research of HPK1 inhibitors, there are mainly two ways: monotherapy and combination therapy. In recent years, HPK1 degraders derived from PROTAC technology have shown promises along with HPK1 inhibitors. It is hopeful that small molecule inhibitors or degraders targeting HPK1 will gain FDA approval for treatment of human diseases in the near future. DATABASES SEARCHED AND INCLUSIVE DATES A rapid survey of literature reports using keyword 'HPK1' in SciFinder® search engine yielded about 180 papers since 2021.
Collapse
Affiliation(s)
- Ying-Hui Yuan
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jia-Ying Mao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Ji-Fan Yue
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Meng-Lan He
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zi Hui
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Hang Yin
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Jianshe Wang
- Drug Discovery, Hangzhou Purple Crystal Pharma, Hangzhou, Zhejiang, China
| | - Xiang-Yang Ye
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Mao J, Zhou L, Wu Y, Wang K, Ye X, Wang T, Yang J, Tong J, Miao Q, Jiang S, Xiao Y, Zhang K. Discovery of 1,2,4-benzotriazine derivatives as new hematopoietic progenitor kinase 1 (HPK1) inhibitors. Bioorg Chem 2025; 156:108158. [PMID: 39826501 DOI: 10.1016/j.bioorg.2025.108158] [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: 10/25/2024] [Revised: 12/26/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Hematopoietic progenitor kinase 1 (HPK1), which negatively regulates immune signaling, has emerged as an attractive small-molecule drug target for tumor immunotherapy. Herein, we report the discovery of the 1,2,4-benzotriazine derivatives as new potent HPK1 inhibitors. Notably, compound A29 exhibited improved HPK1 inhibitory activity relative to compound 1 in the ADP-Glo kinase assay (IC50 = 2.70 and 13.6 nM, respectively). The pronounced inhibitory activity of A29 against downstream p-SLP76 in Jurkat T cells (IC50 = 8.1 nM) as well as the ability to induce the production of interleukin 2 (IL-2) in human peripheral blood mononuclear cells (PBMCs) confirmed its cellular target engagement and immune stimulatory effect. Consistently, this lead compound significantly enhanced T-cell killing ability against murine colon cancer cells CT26 or MC38 in a co-culture system. Furthermore, A29 was efficacious in a CT26 xenograft mouse model alone, and significantly enhanced the antitumor efficacy of an anti-PD-1 antibody. This work provides a promising lead for the development of effective HPK1 inhibitors for tumor immunotherapy.
Collapse
Affiliation(s)
- Jie Mao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lixin Zhou
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yuxing Wu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kaizhen Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiuquan Ye
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tianyu Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jiamei Yang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jun Tong
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qi Miao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Sheng Jiang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Yibei Xiao
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Kuojun Zhang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
5
|
Shields JD, Baker D, Balazs AYS, Bommakanti G, Casella R, Cao S, Cook S, Escobar RA, Fawell S, Gibbons FD, Giblin KA, Goldberg FW, Gosselin E, Grebe T, Hariparsad N, Hatoum-Mokdad H, Howells R, Hughes SJ, Jackson A, Karapa Reddy I, Kettle JG, Lamont GM, Lamont S, Li M, Lill SON, Mele DA, Metrano AJ, Mfuh AM, Morrill LA, Peng B, Pflug A, Proia TA, Rezaei H, Richards R, Richter M, Robbins KJ, San Martin M, Schimpl M, Schuller AG, Sha L, Shen M, Sheppeck JE, Singh M, Stokes S, Song K, Sun Y, Tang H, Wagner DJ, Wang J, Wang Y, Wilson DM, Wu A, Wu C, Wu D, Wu Y, Xu K, Yang Y, Yao T, Ye M, Zhang AX, Zhang H, Zhai X, Zhou Y, Ziegler RE, Grimster NP. Discovery and Optimization of Pyrazine Carboxamide AZ3246, a Selective HPK1 Inhibitor. J Med Chem 2025; 68:4582-4595. [PMID: 39928839 DOI: 10.1021/acs.jmedchem.4c02631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Abstract
Hematopoietic progenitor kinase 1 (HPK1) is a negative regulator of the T cell receptor signaling pathway and is therefore a target of interest for immunooncology. Nonselective HPK1 inhibitors may affect other kinase components of T cell activation, blunting the beneficial impact of enhanced T cell activity that results from HPK1 inhibition itself. Here, we report the discovery of pyrazine carboxamide HPK1 inhibitors and their optimization through structure-based drug design to afford a highly selective HPK1 inhibitor, compound 24 (AZ3246). This compound induces IL-2 secretion in T cells with an EC50 of 90 nM without inhibiting antagonistic kinases, exhibits pharmacokinetic properties consistent with oral dosing, and demonstrates antitumor activity in the EMT6 syngeneic mouse model.
Collapse
Affiliation(s)
- Jason D Shields
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - David Baker
- Discovery Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Amber Y S Balazs
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Gayathri Bommakanti
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Robert Casella
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Shenggen Cao
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Steve Cook
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Randolph A Escobar
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Stephen Fawell
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Francis D Gibbons
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Kathryn A Giblin
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | | | - Eric Gosselin
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Tyler Grebe
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Niresh Hariparsad
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Holia Hatoum-Mokdad
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Rachel Howells
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Samantha J Hughes
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Anne Jackson
- Discovery Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Iswarya Karapa Reddy
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Jason G Kettle
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Gillian M Lamont
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Scott Lamont
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Min Li
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Sten O Nilsson Lill
- Pharmaceutical Sciences, R&D, AstraZeneca, Pepparedsleden 1, SE-431 83 Mölndal, Sweden
| | - Deanna A Mele
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Anthony J Metrano
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Adelphe M Mfuh
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Lucas A Morrill
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Bo Peng
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Alexander Pflug
- Discovery Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Theresa A Proia
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Hadi Rezaei
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Ryan Richards
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Magdalena Richter
- Discovery Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Kevin J Robbins
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Maryann San Martin
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Marianne Schimpl
- Discovery Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Alwin G Schuller
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Li Sha
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Minhui Shen
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - James E Sheppeck
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Meha Singh
- Discovery Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Stephen Stokes
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Kun Song
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Yuanyuan Sun
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Haoran Tang
- Discovery Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - David J Wagner
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Jianyan Wang
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Yanjun Wang
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - David M Wilson
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Allan Wu
- Discovery Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Chengyan Wu
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Dedong Wu
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Ye Wu
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Kevin Xu
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Yue Yang
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Tieguang Yao
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Minwei Ye
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Andrew X Zhang
- Discovery Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Hui Zhang
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Xiang Zhai
- Discovery Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Yanxiao Zhou
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Robert E Ziegler
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Neil P Grimster
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| |
Collapse
|
6
|
Wang Q, Zhu X, Li J, Xu S, Wang A, Zhang X, Wang X, Cai X, Xing H, Liu Y, Liu X, Wang Z, Wang L, Yuan X. HPK1 kinase inhibitor: a sufficient approach to target HPK1 to modulate T cell activation in cancer immunotherapy compared with degraders. Front Immunol 2025; 16:1449106. [PMID: 39981246 PMCID: PMC11839646 DOI: 10.3389/fimmu.2025.1449106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 01/17/2025] [Indexed: 02/22/2025] Open
Abstract
Background Hematopoietic progenitor kinase 1 (HPK1) is a member of the mitogen-activated protein kinase kinase kinase kinase (MAP4K) family. It has been reported that HPK1 negatively regulates the activation of T cells. Several compounds have been developed and tested in clinical trials to target HPK1 for cancer immunotherapy. However, whether kinase inhibition is sufficient to eliminate the immunosuppressive function of HPK1, particularly in T cells, remains elusive. Methods In this study, genetic tools were used to edit the human T lymphocyte cell line Jurkat. The activation of HPK1-null cells, HPK1-wildtype cells and HPK1-kinase-inactive cells was compared through ectopic expression of HPK1 in HPK1 knockout cells or direct HPK1 mutation. Besides genetic validation, a series of compounds that selectively target HPK1 (with or without HPK1-degradation activity) were used to assess the potential scaffold function of HPK1 in regulation of human primary T cell activation and cytotoxic activity. Results and conclusion Augmented T-cell receptor (TCR)-induced activation in HPK1-knockout Jurkat cells was inhibited by complementation of wildtype, but not kinase-dead HPK1. HPK1 K46E-knockin and K46*-knockin Jurkat cells showed comparable levels of enhanced TCR-induced activation compared with control HPK1-wildtype Jurkat cells. Similarly, HPK1 kinase inhibitor (Compound 1) and cereblon-based (CRBN-based) HPK1 degrader (Compound 2) elicited similar degrees of maximum TCR-induced activation in primary human peripheral blood T cells. In summary, the results of this study suggested that HPK1 kinase inhibitor may be sufficient for HPK1 targeting in T cell mediated cancer immunotherapy.
Collapse
Affiliation(s)
- Qin Wang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Xinyi Zhu
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Jing Li
- Department of Medicinal Chemistry, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Sanjia Xu
- Department of Medicinal Chemistry, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Ali Wang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Xinwen Zhang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Xingxing Wang
- Department of Medicinal Chemistry, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Xiaopeng Cai
- Department of Medicinal Chemistry, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Haimei Xing
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Ye Liu
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Xuesong Liu
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Zhiwei Wang
- Department of Medicinal Chemistry, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Lai Wang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
- Research & Clinical Development, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Xi Yuan
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| |
Collapse
|
7
|
Mowat J, Carretero R, Leder G, Aiguabella Font N, Neuhaus R, Berndt S, Günther J, Friberg A, Schäfer M, Briem H, Raschke M, Miyatake Ondozabal H, Buchmann B, Boemer U, Kreft B, Hartung IV, Offringa R. Discovery of BAY-405: An Azaindole-Based MAP4K1 Inhibitor for the Enhancement of T-Cell Immunity against Cancer. J Med Chem 2024; 67:17429-17453. [PMID: 39331123 PMCID: PMC11472321 DOI: 10.1021/acs.jmedchem.4c01325] [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: 06/11/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024]
Abstract
Mitogen-activated protein kinase kinase kinase kinase 1 (MAP4K1) is a serine/threonine kinase that acts as an immune checkpoint downstream of T-cell receptor stimulation. MAP4K1 activity is enhanced by prostaglandin E2 (PGE2) and transforming growth factor beta (TGFβ), immune modulators commonly present in the tumor microenvironment. Therefore, its pharmacological inhibition is an attractive immuno-oncology concept for inducing therapeutic T-cell responses in cancer patients. Here, we describe the systematic optimization of azaindole-based lead compound 1, resulting in the discovery of potent and selective MAP4K1 inhibitor 38 (BAY-405) that displays nanomolar potency in biochemical and cellular assays as well as in vivo exposure after oral dosing. BAY-405 enhances T-cell immunity and overcomes the suppressive effect of PGE2 and TGFβ. Treatment of tumor-bearing mice shows T-cell-dependent antitumor efficacy. MAP4K1 inhibition in conjunction with PD-L1 blockade results in a superior antitumor impact, illustrating the complementarity of the single agent treatments.
Collapse
Affiliation(s)
| | - Rafael Carretero
- Bayer
AG, Pharmaceutical R&D, 13342 Berlin, Germany
- DKFZ-Bayer
Joint Immunotherapeutics Laboratory, German Cancer Research Center, Heidelberg 69120, Germany
| | | | | | - Roland Neuhaus
- DKFZ-Bayer
Joint Immunotherapeutics Laboratory, German Cancer Research Center, Heidelberg 69120, Germany
| | | | | | | | | | - Hans Briem
- Bayer
AG, Pharmaceutical R&D, 13342 Berlin, Germany
| | | | | | | | - Ulf Boemer
- Bayer
AG, Pharmaceutical R&D, 13342 Berlin, Germany
| | | | | | - Rienk Offringa
- DKFZ-Bayer
Joint Immunotherapeutics Laboratory, German Cancer Research Center, Heidelberg 69120, Germany
- Division
of Molecular Oncology of Gastrointestinal Tumors, Department of Surgery, University Hospital Heidelberg, Heidelberg 69120, Germany
| |
Collapse
|
8
|
Tellis JC, Wei B, Siu M, An L, Chan GK, Chen Y, Du X, Gazzard L, Hu B, Kiefer J, Kakiuchi-Kiyota S, Lainchbury M, Linehan JL, Luo X, Malhotra S, Mendonca R, Pang J, Ran Y, Sethuraman V, Seward E, Sneeringer C, Su D, Wang W, Wu P, Moffat JG, Heffron TP, Choo EF, Chan BK. Discovery of GNE-6893, a Potent, Selective, Orally Bioavailable Small Molecule Inhibitor of HPK1. ACS Med Chem Lett 2024; 15:1606-1614. [PMID: 39291002 PMCID: PMC11403726 DOI: 10.1021/acsmedchemlett.4c00319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Hematopoietic progenitor kinase 1 (HPK1) serves a key immunosuppressive role as a negative regulator of T-cell receptor (TCR) signaling. HPK1 loss-of-function is associated with augmentation of immune function and has demonstrated synergy with immune checkpoint inhibitors in syngeneic mouse cancer models. These data offer compelling evidence for the use of selective small molecule inhibitors of HPK1 in cancer immunotherapy. We identified a novel series of isoquinoline HPK1 inhibitors through fragment-based screening that displayed promising levels of biochemical potency and activity in functional cell-based assays. We used structure-based drug design to introduce key selectivity elements while simultaneously addressing pharmacokinetic liabilities. These efforts culminated in a molecule demonstrating subnanomolar biochemical inhibition of HPK1 and strong in vitro augmentation of TCR signaling in primary human T-cells. Further profiling of this molecule revealed excellent kinase selectivity (347/356 kinases <50% inhibition @ 0.1 μM), a favorable in vitro safety profile, and good projected human pharmacokinetics.
Collapse
Affiliation(s)
- John C Tellis
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - BinQing Wei
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Michael Siu
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Le An
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Grace Kayan Chan
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Yong Chen
- Pharmaron Beijing Co., No. 6 Tai He Road, BDA, Beijing 100176, P.R. China
| | - Xiangnan Du
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Lewis Gazzard
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Baihua Hu
- Pharmaron Beijing Co., No. 6 Tai He Road, BDA, Beijing 100176, P.R. China
| | - James Kiefer
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | | | - Michael Lainchbury
- Charles River Laboratories, 8-9 Spire Green, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Jonathan L Linehan
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Xifeng Luo
- Pharmaron Beijing Co., No. 6 Tai He Road, BDA, Beijing 100176, P.R. China
| | - Sushant Malhotra
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Rohan Mendonca
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jodie Pang
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Yinqing Ran
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Vijay Sethuraman
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Eileen Seward
- Charles River Laboratories, 8-9 Spire Green, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Chris Sneeringer
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Dian Su
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Weiru Wang
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Ping Wu
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - John G Moffat
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Timothy P Heffron
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Edna F Choo
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Bryan K Chan
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
9
|
Ou X, Gao G, Habaz IA, Wang Y. Mechanisms of resistance to tyrosine kinase inhibitor-targeted therapy and overcoming strategies. MedComm (Beijing) 2024; 5:e694. [PMID: 39184861 PMCID: PMC11344283 DOI: 10.1002/mco2.694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/27/2024] Open
Abstract
Tyrosine kinase inhibitor (TKI)-targeted therapy has revolutionized cancer treatment by selectively blocking specific signaling pathways crucial for tumor growth, offering improved outcomes with fewer side effects compared with conventional chemotherapy. However, despite their initial effectiveness, resistance to TKIs remains a significant challenge in clinical practice. Understanding the mechanisms underlying TKI resistance is paramount for improving patient outcomes and developing more effective treatment strategies. In this review, we explored various mechanisms contributing to TKI resistance, including on-target mechanisms and off-target mechanisms, as well as changes in the tumor histology and tumor microenvironment (intrinsic mechanisms). Additionally, we summarized current therapeutic approaches aiming at circumventing TKI resistance, including the development of next-generation TKIs and combination therapies. We also discussed emerging strategies such as the use of dual-targeted antibodies and PROteolysis Targeting Chimeras. Furthermore, we explored future directions in TKI-targeted therapy, including the methods for detecting and monitoring drug resistance during treatment, identification of novel targets, exploration of dual-acting kinase inhibitors, application of nanotechnologies in targeted therapy, and so on. Overall, this review provides a comprehensive overview of the challenges and opportunities in TKI-targeted therapy, aiming to advance our understanding of resistance mechanisms and guide the development of more effective therapeutic approaches in cancer treatment.
Collapse
Affiliation(s)
- Xuejin Ou
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Ge Gao
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China HospitalSichuan UniversityChengduChina
| | - Inbar A. Habaz
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonOntarioCanada
| | - Yongsheng Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
10
|
Zhang TT, Yi W, Dong DZ, Ren ZY, Zhang Y, Du F. METTL3-mediated upregulation of FAM135B promotes EMT of esophageal squamous cell carcinoma via regulating the Wnt/β-catenin pathway. Am J Physiol Cell Physiol 2024; 327:C329-C340. [PMID: 38881420 DOI: 10.1152/ajpcell.00529.2023] [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: 10/12/2023] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/18/2024]
Abstract
Family with sequence similarity 135 member B (FAM135B) is a novel driver gene in esophageal squamous cell carcinoma (ESCC). However, little is known regarding its biological functions and mechanisms in ESCC. Here, we identified that the high expression of FAM135B was associated with lymph node metastasis and infiltrating development of ESCC. Elevated FAM135B expression promoted ESCC migration and invasion in vitro and lung metastasis in vivo. Furthermore, epithelial-mesenchymal transition (EMT)-related pathways were enriched in ESCC samples with high levels of FAM135B and FAM135B positively regulated EMT markers. Mechanistically, we observed that FAM135B interacted with the intermediate domain of TRAF2 and NCK-interacting kinase (TNIK), activating the Wnt/β-catenin signaling pathway. The facilitation of TNIK on ESCC migration and invasion was reversed by FAM135B siRNA. In addition, the N6-methyladenosine (m6A) modification positively regulated FAM135B expression, with methyltransferase like 3 (METTL3) acting as its substantial m6A writer. The pro-EMT effects of METTL3 overexpression were reversed by silencing FAM135B. Collectively, these findings illustrate the critical role of ABCDE in ESCC progression and provide new insights into the upstream and downstream mechanisms of FAM135B.NEW & NOTEWORTHY This study reveals for the first time that the novel cancer-related gene, FAM135B, promotes ESCC metastasis both in vitro and in vivo. Besides, we substantiate FAM135B's action on the β-catenin pathway through interacting with TNIK, thereby elucidating the promotional effect of FAM135B on ESCC EMT. Furthermore, we provide initial evidence demonstrating that METTL3-mediated m6A modification upregulates the expression of FAM135B in ESCC cells.
Collapse
Affiliation(s)
- Tong-Tong Zhang
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu/The Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, People's Republic of China
- Medical Research Center, The Third People's Hospital of Chengdu/The Affiliated Hospital of Southwest Jiaotong University, Chengdu, People's Republic of China
| | - Wei Yi
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu/The Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, People's Republic of China
| | - De-Zuo Dong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Zheng-Yun Ren
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu/The Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, People's Republic of China
| | - Yu Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Feng Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), The VIPII Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| |
Collapse
|
11
|
Chen H, Guan X, He C, Lu T, Lin X, Liao X. Current strategies for targeting HPK1 in cancer and the barriers to preclinical progress. Expert Opin Ther Targets 2024; 28:237-250. [PMID: 38650383 DOI: 10.1080/14728222.2024.2344697] [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: 10/26/2023] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
INTRODUCTION Hematopoietic progenitor kinase 1 (HPK1), a 97-kDa serine/threonine Ste20-related protein kinase, functions as an intracellular negative regulator, primarily in hematopoietic lineage cells, where it regulates T cells, B cells, dendritic cells, and other immune cells. Loss of HPK1 kinase activity results in exacerbated cytokine secretion, enhanced T cell signaling, improved viral clearance, and thus increased restraint of tumor growth. These findings highlight HPK1 as a promising target for immuno-oncology treatments, culminating in the advancement of candidate compounds targeting HPK1 to clinical trials by several biotech enterprises. AREAS COVERED Through searching PubMed, Espacenet-patent search, and clinicaltrials.gov, this review provides a comprehensive analysis of HPK1, encompassing its structure and roles in various downstream signaling pathways, the consequences of constitutive activation of HPK1, and potential therapeutic strategies to treat HPK1-driven malignancies. Moreover, the review outlines the patents issued for small molecule inhibitors and clinical investigations of HPK1. EXPERT OPINION To enhance the success of tumor immunotherapy in clinical trials, it is important to develop protein degraders, allosteric inhibitors, and antibody-drug conjugates based on the crystal structure of HPK1, and to explore combination therapy approaches. Although several challenges remain, the development of HPK1 inhibitors display promising in preclinical and clinical studies.
Collapse
Affiliation(s)
- Hui Chen
- State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Science, Tsinghua University, Beijing, China
| | - Xiangna Guan
- State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Science, Tsinghua University, Beijing, China
| | - Chi He
- State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Science, Tsinghua University, Beijing, China
| | - Tingting Lu
- Zhuhai Yufan Biotechnologies Co., Ltd, Zhuhai, Guangdong, China
| | - Xingyu Lin
- Zhuhai Yufan Biotechnologies Co., Ltd, Zhuhai, Guangdong, China
| | - Xuebin Liao
- State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Science, Tsinghua University, Beijing, China
| |
Collapse
|
12
|
Köster KA, Dethlefs M, Duque Escobar J, Oetjen E. Regulation of the Activity of the Dual Leucine Zipper Kinase by Distinct Mechanisms. Cells 2024; 13:333. [PMID: 38391946 PMCID: PMC10886912 DOI: 10.3390/cells13040333] [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: 12/29/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/24/2024] Open
Abstract
The dual leucine zipper kinase (DLK) alias mitogen-activated protein 3 kinase 12 (MAP3K12) has gained much attention in recent years. DLK belongs to the mixed lineage kinases, characterized by homology to serine/threonine and tyrosine kinase, but exerts serine/threonine kinase activity. DLK has been implicated in many diseases, including several neurodegenerative diseases, glaucoma, and diabetes mellitus. As a MAP3K, it is generally assumed that DLK becomes phosphorylated and activated by upstream signals and phosphorylates and activates itself, the downstream serine/threonine MAP2K, and, ultimately, MAPK. In addition, other mechanisms such as protein-protein interactions, proteasomal degradation, dephosphorylation by various phosphatases, palmitoylation, and subcellular localization have been shown to be involved in the regulation of DLK activity or its fine-tuning. In the present review, the diverse mechanisms regulating DLK activity will be summarized to provide better insights into DLK action and, possibly, new targets to modulate DLK function.
Collapse
Affiliation(s)
- Kyra-Alexandra Köster
- Department of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany; (K.-A.K.); (M.D.)
- DZHK Standort Hamburg, Kiel, Lübeck, Germany;
| | - Marten Dethlefs
- Department of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany; (K.-A.K.); (M.D.)
- DZHK Standort Hamburg, Kiel, Lübeck, Germany;
| | - Jorge Duque Escobar
- DZHK Standort Hamburg, Kiel, Lübeck, Germany;
- University Center of Cardiovascular Science, Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Elke Oetjen
- Department of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany; (K.-A.K.); (M.D.)
- DZHK Standort Hamburg, Kiel, Lübeck, Germany;
- Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany
| |
Collapse
|
13
|
Zhang J, Ren Z, Hu Y, Shang S, Wang R, Ma J, Zhang Z, Wu M, Wang F, Yu J, Chen D. High HPK1 +PD-1 +TIM-3 +CD8 + T cells infiltration predicts poor prognosis to immunotherapy in NSCLC patients. Int Immunopharmacol 2024; 127:111363. [PMID: 38101218 DOI: 10.1016/j.intimp.2023.111363] [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: 09/20/2023] [Revised: 11/21/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
At present the efficacy of immune checkpoint inhibitors (ICIs) remains limited. The lack of responsiveness in certain patients may be attributed to CD8+ T cell exhaustion within the tumor microenvironment (TME). Hematopoietic progenitor kinase 1 (HPK1) has been identified as a mediator of T cell dysfunction, leading to our hypothesis that HPK1 positive exhausted CD8+ T cells could serve as a predictor for ICIs' efficacy in NSCLC patients, and potentially indicate key cellular subset causing ICIs resistance. Here, we retrospectively collected tumor tissue samples from 36 NSCLC patients who underwent first-line immunotherapy. Using multiplex immunohistochemistry, we visualized various PD-1+CD8+ T cell subsets and explore biomarkers for response. The analysis endpoints included overall response rate (ORR), progression free survival (PFS), and overall survival (OS), correlating them with levels of cell infiltration or effective density. We found that the proportion of PD-1+CD8+ T cell subsets did not align with predictions for ORR, PFS, and OS. Conversely, a high infiltration of HPK1+PD-1+TIM-3+CD8+ T cells was identified as an independent risk factor for both PFS (P = 0.019) and OS (P = 0.03). These cells were found to express the highest levels of Granzyme B, and the secretion of Granzyme B in CD8+ T cell subsets was related to TCF-1. In conclusion, these data suggest that a high infiltration of HPK1+PD-1+TIM-3+CD8+ T cells correlates with poor clinical outcomes in NSCLC patients receiving immunotherapy. These cells may represent terminally exhausted T cells that fail to respond to ICIs, thereby laying the groundwork for the potential integration of HPK1 inhibitors with immunotherapy to enhance treatment strategy.
Collapse
Affiliation(s)
- Jingxin Zhang
- Shandong University Cancer Center, Jinan, Shandong, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ziyuan Ren
- Shandong University Cancer Center, Jinan, Shandong, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yun Hu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shijie Shang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ruiyang Wang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jiachun Ma
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zengfu Zhang
- Shandong University Cancer Center, Jinan, Shandong, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Meng Wu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Fei Wang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinming Yu
- Shandong University Cancer Center, Jinan, Shandong, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| | - Dawei Chen
- Shandong University Cancer Center, Jinan, Shandong, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
14
|
Zhang J, Li Y, Tang H, Zhou Q, Tong L, Ding J, Xie H, Xiong B, Liu T. Design and synthesis of 1H-pyrazolo[3,4-d]pyrimidine derivatives as hematopoietic progenitor kinase 1 (HPK1) inhibitors. Bioorg Chem 2023; 140:106811. [PMID: 37659145 DOI: 10.1016/j.bioorg.2023.106811] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/07/2023] [Accepted: 08/22/2023] [Indexed: 09/04/2023]
Abstract
Despite immune checkpoint inhibitors' tremendous success in the treatment of tumors, the moderate response rate limits their widespread use. Hematopoietic progenitor kinase 1 (HPK1) is served as an essential negative regulator of T-cell receptor, which has been identified as a promising target for enhancing antitumor immunity. However, the development of a selective HPK1 inhibitor is still challenging. Herein, we reported a novel series of 1H-pyrazolo[3,4-d]pyrimidine derivatives as HPK1 inhibitors by structure-based rational design. The optimal compound 10n significantly inhibited HPK1 with an IC50 value of 29.0 nM and the phosphorylation of SLP76 at a concentration as low as 0.1 μM. Furthermore, compound 10n exhibited good selectivity over a panel of 25 kinases, including GLK from the same MAP4K family. Together, the current study provided a novel, potent, and selective HPK1 inhibitor, acting as a lead compound for the future development of cancer immunotherapy.
Collapse
Affiliation(s)
- Junjie Zhang
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yan Li
- Division of Antitumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Haotian Tang
- Division of Antitumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Qianqian Zhou
- Division of Antitumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China; Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, PR China
| | - Linjiang Tong
- Division of Antitumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Jian Ding
- Division of Antitumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China; Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, PR China
| | - Hua Xie
- Division of Antitumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, PR China.
| | - Bing Xiong
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Tongchao Liu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China.
| |
Collapse
|
15
|
Toure M, Johnson T, Li B, Schmidt R, Ma H, Neagu C, Lopez AU, Wang Y, Guler S, Xiao Y, Henkes R, Ho K, Zhang S, Chu CL, Gundra UM, Porichis F, Li L, Maurer CK, Fang Z, Musil D, DiPoto M, Friis E, Jones R, Jones C, Cummings J, Chekler E, Tanzer EM, Huck B, Sherer B. Discovery of quinazoline HPK1 inhibitors with high cellular potency. Bioorg Med Chem 2023; 92:117423. [PMID: 37531921 DOI: 10.1016/j.bmc.2023.117423] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
Hematopoietic progenitor kinase 1 (HPK1) is regarded as a highly validated target in pre-clinical immune oncology. HPK1 has been described as regulating multiple critical signaling pathway in both adaptive and innate cells. In support of this role, HPK1 KO T cells show enhanced sensitivity to TCR activation and HPK1 KO mice display enhanced anti-tumor activity. Taken together, inhibition of HPK1 has the potential to induce enhanced anti-tumor immune response. Herein, we described the discovery of highly potent HPK1 inhibitors starting form a weak HTS hit. Using a structure-based drug design, HPK1 inhibitors exhibiting excellent cellular single-digit nanomolar potency in both proximal (pSLP76) and distal (IL-2) biomarkers along with sustained elevation of IL-2 cytokine secretion were discovered.
Collapse
Affiliation(s)
- Momar Toure
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States.
| | - Theresa Johnson
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Bin Li
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Ralf Schmidt
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Hong Ma
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Constantin Neagu
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Andrea Unzue Lopez
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Yanping Wang
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Satenig Guler
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - YuFang Xiao
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Renate Henkes
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Kevin Ho
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Susan Zhang
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Chia Lin Chu
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Uma Mahesh Gundra
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Filippos Porichis
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Long Li
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Christine Katharina Maurer
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Zhizhou Fang
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Djordje Musil
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Maria DiPoto
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Emily Friis
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Reinaldo Jones
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Christopher Jones
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - James Cummings
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Eugene Chekler
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Eva Maria Tanzer
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Bayard Huck
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| | - Brian Sherer
- Discovery & Development Technologies, Medicinal Chemistry, EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, United States
| |
Collapse
|
16
|
Zhou L, Ye X, Wang K, Shen H, Wang T, Zhang X, Jiang S, Xiao Y, Zhang K. Discovery of diaminotriazine carboxamides as potent inhibitors of hematopoetic progenitor kinase 1. Bioorg Chem 2023; 138:106682. [PMID: 37339563 DOI: 10.1016/j.bioorg.2023.106682] [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: 11/06/2022] [Revised: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 06/22/2023]
Abstract
Hematopoietic progenitor kinase 1 (HPK1), a member of mitogen-activated protein kinase kinase kinase kinase (MAP4K) family of Ste20 serine/threonine kinases, is a negative regulator of T-cell receptor (TCR) signaling. Inactivating HPK1 kinase has been reported to be sufficient to elicit antitumor immune response. Therefore, HPK1 has attracted much attention as a promising target for tumor immunotherapy. A few of HPK1 inhibitors have been reported, and none of them have been approved for clinical applications. Hence, more effective HPK1 inhibitors are needed. Herein, a series of structurally novel diaminotriazine carboxamides were rationally designed, synthesized and evaluated for their inhibitory activity against HPK1 kinase. Most of them exhibited potent inhibitory potency against HPK1 kinase. In particular, compound 15b showed more robust HPK1 inhibitory activity than that of 11d developed by Merck in kinase activity assay (IC50 = 3.1 and 8.2 nM, respectively). The significant inhibitory potency against SLP76 phosphorylation in Jurkat T cells further confirmed the efficacy of compound 15b. In human peripheral blood mononuclear cell (PBMC) functional assays, compound 15b more significantly induced the production of interleukin 2 (IL-2) and interferon γ (IFN-γ) relative to 11d. Furthermore, 15b alone or in combination with anti-PD-1 antibodies showed potent in vivo antitumor efficacy in MC38 tumor-bearing mice. Compound 15b represents a promising lead for the development of effective HPK1 small-molecule inhibitors.
Collapse
Affiliation(s)
- Lixin Zhou
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiuquan Ye
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kaizhen Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Hongtao Shen
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tianyu Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiangyu Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Sheng Jiang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Yibei Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Kuojun Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
17
|
Wang H, Moniruzzaman R, Li L, Ji B, Liu Y, Zuo X, Abbasgholizadeh R, Zhao J, Liu G, Wang R, Tang H, Sun R, Su X, Tan TH, Maitra A, Wang H. Hematopoietic progenitor kinase 1 inhibits the development and progression of pancreatic intraepithelial neoplasia. J Clin Invest 2023; 133:e163873. [PMID: 37140994 PMCID: PMC10266776 DOI: 10.1172/jci163873] [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: 08/03/2022] [Accepted: 05/02/2023] [Indexed: 05/05/2023] Open
Abstract
Ras plays an essential role in the development of acinar-to-ductal metaplasia (ADM) and pancreatic ductal adenocarcinoma (PDAC). However, mutant Kras is an inefficient driver for PDAC development. The mechanisms of the switching from low Ras activity to high Ras activity that are required for development and progression of pancreatic intraepithelial neoplasias (PanINs) are unclear. In this study, we found that hematopoietic progenitor kinase 1 (HPK1) was upregulated during pancreatic injury and ADM. HPK1 interacted with the SH3 domain and phosphorylated Ras GTPase-activating protein (RasGAP) and upregulated RasGAP activity. Using transgenic mouse models of HPK1 or M46, a kinase-dead mutant of HPK1, we showed that HPK1 inhibited Ras activity and its downstream signaling and regulated acinar cell plasticity. M46 promoted the development of ADM and PanINs. Expression of M46 in KrasG12D Bac mice promoted the infiltration of myeloid-derived suppressor cells and macrophages, inhibited the infiltration of T cells, and accelerated the progression of PanINs to invasive and metastatic PDAC, while HPK1 attenuated mutant Kras-driven PanIN progression. Our results showed that HPK1 plays an important role in ADM and the progression of PanINs by regulating Ras signaling. Loss of HPK1 kinase activity promotes an immunosuppressive tumor microenvironment and accelerates the progression of PanINs to PDAC.
Collapse
Affiliation(s)
- Hua Wang
- Department of Gastrointestinal Medical Oncology and
| | - Rohan Moniruzzaman
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lei Li
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Baoan Ji
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Yi Liu
- Department of Gastrointestinal Medical Oncology and
| | | | - Reza Abbasgholizadeh
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jun Zhao
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Guangchao Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ruiqi Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Ryan Sun
- Department of Biostatistics, and
| | - Xiaoping Su
- Advanced Technology Genomics Core
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Translational Molecular Pathology and
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Translational Molecular Pathology and
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
18
|
Singh SK, Roy R, Kumar S, Srivastava P, Jha S, Rana B, Rana A. Molecular Insights of MAP4K4 Signaling in Inflammatory and Malignant Diseases. Cancers (Basel) 2023; 15:cancers15082272. [PMID: 37190200 PMCID: PMC10136566 DOI: 10.3390/cancers15082272] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Mitogen-activated protein kinase (MAPK) cascades are crucial in extracellular signal transduction to cellular responses. The classical three-tiered MAPK cascades include signaling through MAP kinase kinase kinase (MAP3K) that activates a MAP kinase kinase (MAP2K), which in turn induces MAPK activation and downstream cellular responses. The upstream activators of MAP3K are often small guanosine-5'-triphosphate (GTP)-binding proteins, but in some pathways, MAP3K can be activated by another kinase, which is known as a MAP kinase kinase kinase kinase (MAP4K). MAP4K4 is one of the widely studied MAP4K members, known to play a significant role in inflammatory, cardiovascular, and malignant diseases. The MAP4K4 signal transduction plays an essential role in cell proliferation, transformation, invasiveness, adhesiveness, inflammation, stress responses, and cell migration. Overexpression of MAP4K4 is frequently reported in many cancers, including glioblastoma, colon, prostate, and pancreatic cancers. Besides its mainstay pro-survival role in various malignancies, MAP4K4 has been implicated in cancer-associated cachexia. In the present review, we discuss the functional role of MAP4K4 in malignant/non-malignant diseases and cancer-associated cachexia and its possible use in targeted therapy.
Collapse
Affiliation(s)
- Sunil Kumar Singh
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Ruchi Roy
- UICentre for Drug Discovery, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sandeep Kumar
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
- University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Piush Srivastava
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Saket Jha
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Basabi Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
- University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
- University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
19
|
Ye Q, Liu K, Ye HF, Pan J, Sokolsky A, Wang A, Zhang K, Hummel JR, Kong L, Behshad E, He X, Conlen P, Stump K, Ye M, Diamond S, Covington M, Yeleswaram S, Atasoylu O, Vechorkin O, Yao W. Discovery of Pyrazolopyridine Derivatives as HPK1 Inhibitors. ACS Med Chem Lett 2023; 14:5-10. [PMID: 36655125 PMCID: PMC9841581 DOI: 10.1021/acsmedchemlett.2c00238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
In spite of the great success of immune checkpoint inhibitors in immune-oncology therapy, an urgent need still exists to identify alternative approaches to broaden the scope of therapeutic coverage. Hematopoietic progenitor kinase 1 (HPK1), also known as MAP4K1, functions as a negative regulator of activation signals generated by the T cell antigen receptor. Herein we report the discovery of novel pyrazolopyridine derivatives as selective inhibitors of HPK1. The structure-activity relationship campaign led to the discovery of compound 16, which has shown promising enzymatic and cellular potency with encouraging kinome selectivity. The outstanding pharmacokinetic profiles of 16 in rats and monkeys supported further evaluations of its efficacy and safety in preclinical models.
Collapse
Affiliation(s)
- Qinda Ye
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Kai Liu
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Hai-Fen Ye
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Jun Pan
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Alexander Sokolsky
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Anlai Wang
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Ke Zhang
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Joshua R. Hummel
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Ling Kong
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Elham Behshad
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Xin He
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Patricia Conlen
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Kristine Stump
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Min Ye
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Sharon Diamond
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Maryanne Covington
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Swamy Yeleswaram
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Onur Atasoylu
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Oleg Vechorkin
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Wenqing Yao
- Incyte Research Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| |
Collapse
|
20
|
Sokolsky A, Vechorkin O, Hummel JR, Styduhar ED, Wang A, Nguyen MH, Ye HF, Liu K, Zhang K, Pan J, Ye Q, Atasoylu O, Behshad E, He X, Conlen P, Stump K, Ye M, Diamond S, Covington M, Yeleswaram S, Yao W. Potent and Selective Biaryl Amide Inhibitors of Hematopoietic Progenitor Kinase 1 (HPK1). ACS Med Chem Lett 2023; 14:116-122. [PMID: 36655134 PMCID: PMC9841582 DOI: 10.1021/acsmedchemlett.2c00241] [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: 05/20/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Herein we report the discovery of a novel biaryl amide series as selective inhibitors of hematopoietic protein kinase 1 (HPK1). Structure-activity relationship development, aided by molecular modeling, identified indazole 5b as a core for further exploration because of its outstanding enzymatic and cellular potency coupled with encouraging kinome selectivity. Late-stage manipulation of the right-hand aryl and amine moieties surmounted issues of selectivity over TRKA, MAP4K2, and STK4 as well as generating compounds with balanced in vitro ADME profiles and promising pharmacokinetics.
Collapse
Affiliation(s)
- Alexander Sokolsky
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Oleg Vechorkin
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Joshua R. Hummel
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Evan D. Styduhar
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Anlai Wang
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Minh H. Nguyen
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Hai Fen Ye
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Kai Liu
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Ke Zhang
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Jun Pan
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Qinda Ye
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Onur Atasoylu
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Elham Behshad
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Xin He
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Patricia Conlen
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Kristine Stump
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Min Ye
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Sharon Diamond
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Maryanne Covington
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Swamy Yeleswaram
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| | - Wenqing Yao
- Incyte Research
Institute, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United
States
| |
Collapse
|
21
|
Zhou L, Wang T, Zhang K, Zhang X, Jiang S. The development of small-molecule inhibitors targeting HPK1. Eur J Med Chem 2022; 244:114819. [DOI: 10.1016/j.ejmech.2022.114819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/25/2022]
|
22
|
Hunter JE, Campbell AE, Kerridge S, Fraser C, Hannaway NL, Luli S, Ivanova I, Brownridge PJ, Coxhead J, Taylor L, Leary P, Hasoon MSR, Eyers CE, Perkins ND. Up-regulation of the PI3K/AKT and RHO/RAC/PAK signalling pathways in CHK1 inhibitor resistant Eµ-Myc lymphoma cells. Biochem J 2022; 479:2131-2151. [PMID: 36240067 PMCID: PMC9704644 DOI: 10.1042/bcj20220103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 12/14/2022]
Abstract
The development of resistance and the activation of bypass pathway signalling represents a major problem for the clinical application of protein kinase inhibitors. While investigating the effect of either a c-Rel deletion or RelAT505A phosphosite knockin on the Eµ-Myc mouse model of B-cell lymphoma, we discovered that both NF-κB subunit mutations resulted in CHK1 inhibitor resistance, arising from either loss or alteration of CHK1 activity, respectively. However, since Eµ-Myc lymphomas depend on CHK1 activity to cope with high levels of DNA replication stress and consequent genomic instability, it was not clear how these mutant NF-κB subunit lymphomas were able to survive. To understand these survival mechanisms and to identify potential compensatory bypass signalling pathways in these lymphomas, we applied a multi-omics strategy. With c-Rel-/- Eµ-Myc lymphomas we observed high levels of Phosphatidyl-inositol 3-kinase (PI3K) and AKT pathway activation. Moreover, treatment with the PI3K inhibitor Pictilisib (GDC-0941) selectively inhibited the growth of reimplanted c-Rel-/- and RelAT505A, but not wild type (WT) Eµ-Myc lymphomas. We also observed up-regulation of a RHO/RAC pathway gene expression signature in both Eµ-Myc NF-κB subunit mutation models. Further investigation demonstrated activation of the RHO/RAC effector p21-activated kinase (PAK) 2. Here, the PAK inhibitor, PF-3758309 successfully overcame resistance of RelAT505A but not WT lymphomas. These findings demonstrate that up-regulation of multiple bypass pathways occurs in CHK1 inhibitor resistant Eµ-Myc lymphomas. Consequently, drugs targeting these pathways could potentially be used as either second line or combinatorial therapies to aid the successful clinical application of CHK1 inhibitors.
Collapse
Affiliation(s)
- Jill E. Hunter
- Newcastle University Biosciences Institute, Wolfson Childhood Cancer Research Centre, Newcastle University, Level 6, Herschel Building, Brewery Lane, Newcastle upon Tyne NE1 7RU, U.K
| | - Amy E. Campbell
- Centre for Proteome Research, Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, U.K
| | - Scott Kerridge
- Newcastle University Biosciences Institute, Wolfson Childhood Cancer Research Centre, Newcastle University, Level 6, Herschel Building, Brewery Lane, Newcastle upon Tyne NE1 7RU, U.K
| | - Callum Fraser
- Newcastle University Biosciences Institute, Wolfson Childhood Cancer Research Centre, Newcastle University, Level 6, Herschel Building, Brewery Lane, Newcastle upon Tyne NE1 7RU, U.K
| | - Nicola L. Hannaway
- Newcastle University Biosciences Institute, Wolfson Childhood Cancer Research Centre, Newcastle University, Level 6, Herschel Building, Brewery Lane, Newcastle upon Tyne NE1 7RU, U.K
| | - Saimir Luli
- Newcastle University Clinical and Translational Research Institute, Preclinical In Vivo Imaging (PIVI), Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE2 4HH, U.K
| | - Iglika Ivanova
- Newcastle University Biosciences Institute, Wolfson Childhood Cancer Research Centre, Newcastle University, Level 6, Herschel Building, Brewery Lane, Newcastle upon Tyne NE1 7RU, U.K
| | - Philip J. Brownridge
- Centre for Proteome Research, Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, U.K
| | - Jonathan Coxhead
- Newcastle University Biosciences Institute, Wolfson Childhood Cancer Research Centre, Newcastle University, Level 6, Herschel Building, Brewery Lane, Newcastle upon Tyne NE1 7RU, U.K
| | - Leigh Taylor
- Newcastle University Biosciences Institute, Wolfson Childhood Cancer Research Centre, Newcastle University, Level 6, Herschel Building, Brewery Lane, Newcastle upon Tyne NE1 7RU, U.K
| | - Peter Leary
- Bioinformatics Support Unit, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE2 4HH, U.K
| | - Megan S. R. Hasoon
- Bioinformatics Support Unit, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE2 4HH, U.K
| | - Claire E. Eyers
- Centre for Proteome Research, Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, U.K
| | - Neil D. Perkins
- Newcastle University Biosciences Institute, Wolfson Childhood Cancer Research Centre, Newcastle University, Level 6, Herschel Building, Brewery Lane, Newcastle upon Tyne NE1 7RU, U.K
| |
Collapse
|
23
|
Zhu Q, Chen N, Tian X, Zhou Y, You Q, Xu X. Hematopoietic Progenitor Kinase 1 in Tumor Immunology: A Medicinal Chemistry Perspective. J Med Chem 2022; 65:8065-8090. [PMID: 35696642 DOI: 10.1021/acs.jmedchem.2c00172] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Hematopoietic progenitor kinase 1 (HPK1), a hematopoietic cell-restricted member of the serine/threonine Ste20-related protein kinases, is a negative regulator of the T cell receptor, B cell receptor, and dendritic cells. Loss of HPK1 kinase function increases cytokine secretion and enhances T cell signaling, virus clearance, and tumor growth inhibition. Therefore, HPK1 is considered a promising target for tumor immunotherapy. Several HPK1 inhibitors have been reported to regulate T cell function. In addition, HPK1-targeting PROTACs, which can induce the degradation of HPK1, have also been developed. Here, we provide an overview of research concerning HPK1 protein structure, function, and inhibitors and propose perspectives and insights for the future development of agents targeting HPK1.
Collapse
Affiliation(s)
- Qiangsheng Zhu
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Nannan Chen
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xinjian Tian
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yeling Zhou
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - QiDong You
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoli Xu
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
24
|
Malchow S, Korepanova A, Panchal SC, McClure RA, Longenecker KL, Qiu W, Zhao H, Cheng M, Guo J, Klinge KL, Trusk P, Pratt SD, Li T, Kurnick MD, Duan L, Shoemaker AR, Gopalakrishnan SM, Warder SE, Shotwell JB, Lai A, Sun C, Osuma AT, Pappano WN. The HPK1 Inhibitor A-745 Verifies the Potential of Modulating T Cell Kinase Signaling for Immunotherapy. ACS Chem Biol 2022; 17:556-566. [PMID: 35188729 DOI: 10.1021/acschembio.1c00819] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hematopoietic progenitor kinase 1 (HPK1) is an MAP4K family member within the Ste20-like serine/threonine branch of the kinome. HPK1 expression is limited to hematopoietic cells and has a predominant role as a negative regulator of T cell function. Because of the central/dominant role in negatively regulating T cell function, HPK1 has long been in the center of interest as a potential pharmacological target for immune therapy. The development of a small molecule HPK1 inhibitor remains challenging because of the need for high specificity relative to other kinases, including additional MAP4K family members, that are required for efficient immune cell activation. Here, we report the identification of the selective and potent HPK1 chemical probe, A-745. In unbiased cellular kinase-binding assays, A-745 demonstrates an excellent cellular selectivity binding profile within pharmacologically relevant concentrations. This HPK1 selectivity translates to an in vitro immune cell activation phenotype reminiscent of Hpk1-deficient and Hpk1-kinase-dead T cells, including augmented proliferation and cytokine production. The results from this work give a path forward for further developmental efforts to generate additional selective and potent small molecule HPK1 inhibitors with the pharmacological properties for immunotherapy.
Collapse
Affiliation(s)
- Sven Malchow
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Alla Korepanova
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Sanjay C. Panchal
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Ryan A. McClure
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | | | - Wei Qiu
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Hongyu Zhao
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Min Cheng
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Jun Guo
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Kelly L. Klinge
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Patricia Trusk
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Steven D. Pratt
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Tao Li
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Matthew D. Kurnick
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Lishu Duan
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Alex R. Shoemaker
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | | | - Scott E. Warder
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - J. Brad Shotwell
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Albert Lai
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Chaohong Sun
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Augustine T. Osuma
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - William N. Pappano
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| |
Collapse
|
25
|
Wang J, Kimura E, Mongan M, Xia Y. Genetic Control of MAP3K1 in Eye Development and Sex Differentiation. Cells 2021; 11:cells11010034. [PMID: 35011600 PMCID: PMC8750206 DOI: 10.3390/cells11010034] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/03/2021] [Accepted: 12/21/2021] [Indexed: 01/11/2023] Open
Abstract
The MAP3K1 is responsible for transmitting signals to activate specific MAP2K-MAPK cascades. Following the initial biochemical characterization, genetic mouse models have taken center stage to elucidate how MAP3K1 regulates biological functions. To that end, mice were generated with the ablation of the entire Map3k1 gene, the kinase domain coding sequences, or ubiquitin ligase domain mutations. Analyses of the mutants identify diverse roles that MAP3K1 plays in embryonic survival, maturation of T/B cells, and development of sensory organs, including eye and ear. Specifically in eye development, Map3k1 loss-of-function was found to be autosomal recessive for congenital eye abnormalities, but became autosomal dominant in combination with Jnk and RhoA mutations. Additionally, Map3k1 mutation increased eye defects with an exposure to environmental agents such as dioxin. Data from eye developmental models reveal the nexus role of MAP3K1 in integrating genetic and environmental signals to control developmental activities. Here, we focus the discussions on recent advances in understanding the signaling mechanisms of MAP3K1 in eye development in mice and in sex differentiation from human genomics findings. The research works featured here lead to a deeper understanding of the in vivo signaling network, the mechanisms of gene-environment interactions, and the relevance of this multifaceted protein kinase in disease etiology and pathogenesis.
Collapse
Affiliation(s)
| | | | | | - Ying Xia
- Correspondence: ; Tel.: +1-513-558-0371
| |
Collapse
|
26
|
He TS, Huang J, Chen T, Zhang Z, Cai K, Yu J, Xu LG. The Kinase MAP4K1 Inhibits Cytosolic RNA-Induced Antiviral Signaling by Promoting Proteasomal Degradation of TBK1/IKKε. Microbiol Spectr 2021; 9:e0145821. [PMID: 34908452 PMCID: PMC8672915 DOI: 10.1128/spectrum.01458-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/15/2021] [Indexed: 11/30/2022] Open
Abstract
TANK-binding kinase 1 (TBK1)/IκB kinase-ε (IKKε) mediates robust production of type I interferons (IFN-I) and proinflammatory cytokines in response to acute viral infection. However, excessive or prolonged production of IFN-I is harmful and even fatal to the host by causing autoimmune disorders. In this study, we identified mitogen-activated protein kinase kinase kinase kinase 1 (MAP4K1) as a negative regulator in the RIG-I-like receptor (RLR) signaling pathway. MAP4K1, a member of Ste20-like serine/threonine kinases, was previously known as a prominent regulator in adaptive immunity by downregulating T-cell receptor (TCR) signaling and B-cell receptor (BCR) signaling. However, its role in regulating antiviral innate immune signaling is still unclear. This study reports an undiscovered role of MAP4K1, which inhibits RLR signaling by targeting TBK1/IKKε for proteasomal degradation via the ubiquitin ligase DTX4. We initially identify MAP4K1 as an interacting partner of TBK1 by yeast two-hybrid screens and subsequently investigate its function in RLR-mediated antiviral signaling pathways. Overexpression of MAP4K1 significantly inhibits RNA virus-triggered activation of IFN-β and the production of proinflammatory cytokines. Consistently, knockdown or knockout experiments show opposite effects. Furthermore, MAP4K1 promotes the degradation of TBK1/IKKε by K48-linked ubiquitination via DTX4. Knockdown of DTX4 abrogated the ubiquitination and degradation of TBK1/IKKε. Collectively, our results identify that MAP4K1 acts as a negative regulator in antiviral innate immunity by targeting TBK1/IKKε, discover a novel TBK1 inhibitor, and extend a novel functional role of MAP4K1 in immunity. IMPORTANCE TANK-binding kinase 1 (TBK1)/IκB kinase-ε (IKKε) mediates robust production of type I interferons (IFN-I) and proinflammatory cytokines to restrict the spread of invading viruses. However, excessive or prolonged production of IFN-I is harmful to the host by causing autoimmune disorders. In this study, we identified that mitogen-activated protein kinase kinase kinase kinase 1 (MAP4K1) is a negative regulator in the RLR signaling pathway. Notably, MAP4K1 promotes the degradation of TBK1/IKKε by K48-linked ubiquitination via the ubiquitin ligase DTX4, leading to the negative regulation of the IFN signaling pathway. Previous studies showed that MAP4K1 has a pivotal function in adaptive immune responses. This study identifies that MAP4K1 also plays a vital role in innate immunity and outlines a novel mechanism by which the IFN signaling pathway is tightly controlled to avoid excessive inflammation. Our study documents a novel TBK1 inhibitor, which serves as a potential therapeutic target for autoimmune diseases, and elucidated a significant function for MAP4K1 linked to innate immunity in addition to subsequent adaptive immunity.
Collapse
Affiliation(s)
- Tian-Sheng He
- College of Life Science, Jiangxi Normal University, Nanchang, China
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Jingping Huang
- College of Life Science, Jiangxi Normal University, Nanchang, China
| | - Tian Chen
- College of Life Science, Jiangxi Normal University, Nanchang, China
| | - Zhi Zhang
- College of Life Science, Jiangxi Normal University, Nanchang, China
| | - Kuntai Cai
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Jingge Yu
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Liang-Guo Xu
- College of Life Science, Jiangxi Normal University, Nanchang, China
| |
Collapse
|
27
|
Dale B, Cheng M, Park KS, Kaniskan HÜ, Xiong Y, Jin J. Advancing targeted protein degradation for cancer therapy. Nat Rev Cancer 2021; 21:638-654. [PMID: 34131295 PMCID: PMC8463487 DOI: 10.1038/s41568-021-00365-x] [Citation(s) in RCA: 363] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/23/2021] [Indexed: 02/05/2023]
Abstract
The human proteome contains approximately 20,000 proteins, and it is estimated that more than 600 of them are functionally important for various types of cancers, including nearly 400 non-enzyme proteins that are challenging to target by traditional occupancy-driven pharmacology. Recent advances in the development of small-molecule degraders, including molecular glues and heterobifunctional degraders such as proteolysis-targeting chimeras (PROTACs), have made it possible to target many proteins that were previously considered undruggable. In particular, PROTACs form a ternary complex with a hijacked E3 ubiquitin ligase and a target protein, leading to polyubiquitination and degradation of the target protein. The broad applicability of this approach is facilitated by the flexibility of individual E3 ligases to recognize different substrates. The vast majority of the approximately 600 human E3 ligases have not been explored, thus presenting enormous opportunities to develop degraders that target oncoproteins with tissue, tumour and subcellular selectivity. In this Review, we first discuss the molecular basis of targeted protein degradation. We then offer a comprehensive account of the most promising degraders in development as cancer therapies to date. Lastly, we provide an overview of opportunities and challenges in this exciting field.
Collapse
Affiliation(s)
- Brandon Dale
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meng Cheng
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kwang-Su Park
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - H Ümit Kaniskan
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yue Xiong
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Cullgen Inc., San Diego, CA, USA.
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
28
|
Linney ID, Kaila N. Inhibitors of immuno-oncology target HPK1 - a patent review (2016 to 2020). Expert Opin Ther Pat 2021; 31:893-910. [PMID: 33956554 DOI: 10.1080/13543776.2021.1924671] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Hematopoietic progenitor kinase (HPK1), a serine/threonine kinase, which is primarily expressed in hematopoietic cells is a negative regulator of T-cell receptor and B cell signaling. Studies using genetic disruption of HPK1 function show enhanced T-cell signaling, cytokine production, and in vivo tumor growth inhibition. This profile of enhanced immune response highlights small molecule inhibition of HPK1 as an attractive approach for the immunotherapy of cancer.Areas covered: This article summarizes the biological rationale for the inhibition of HPK1 as a potential adjunct to the current immuno-oncology (IO) therapies. The article primarily discloses the current state of development of HPK1 inhibitors.Expert Opinion: The rapid increase in the identification of small molecule inhibitors of HPK1 should translate into a fuller understanding of the role of HPK1 inhibition in the IO setting. This understanding will be of huge importance in determining whether HPK1 inhibition alone will be sufficient for tumor growth inhibition or if combination with current IO therapies will be required.
Collapse
Affiliation(s)
- Ian D Linney
- Medicinal Chemistry, Charles River, Chesterford Park Research Park, Saffron Walden, United Kingdom
| | - Neelu Kaila
- Medicinal Chemistry, Nimbus Therapeutics, Cambridge, MA, USA
| |
Collapse
|
29
|
Vara BA, Levi SM, Achab A, Candito DA, Fradera X, Lesburg CA, Kawamura S, Lacey BM, Lim J, Methot JL, Xu Z, Xu H, Smith DM, Piesvaux JA, Miller JR, Bittinger M, Ranganath SH, Bennett DJ, DiMauro EF, Pasternak A. Discovery of Diaminopyrimidine Carboxamide HPK1 Inhibitors as Preclinical Immunotherapy Tool Compounds. ACS Med Chem Lett 2021; 12:653-661. [PMID: 33859804 PMCID: PMC8040257 DOI: 10.1021/acsmedchemlett.1c00096] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/15/2021] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic progenitor kinase 1 (HPK1), a serine/threonine kinase, is a negative immune regulator of T cell receptor (TCR) and B cell signaling that is primarily expressed in hematopoietic cells. Accordingly, it has been reported that HPK1 loss-of-function in HPK1 kinase-dead syngeneic mouse models shows enhanced T cell signaling and cytokine production as well as tumor growth inhibition in vivo, supporting its value as an immunotherapeutic target. Herein, we present the structurally enabled discovery of novel, potent, and selective diaminopyrimidine carboxamide HPK1 inhibitors. The key discovery of a carboxamide moiety was essential for enhanced enzyme inhibitory potency and kinome selectivity as well as sustained elevation of cellular IL-2 production across a titration range in human peripheral blood mononuclear cells. The elucidation of structure-activity relationships using various pendant amino ring systems allowed for the identification of several small molecule type-I inhibitors with promising in vitro profiles.
Collapse
Affiliation(s)
- Brandon A. Vara
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Samuel M. Levi
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Abdelghani Achab
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - David A. Candito
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Xavier Fradera
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Charles A. Lesburg
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Shuhei Kawamura
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Brian M. Lacey
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Jongwon Lim
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Joey L. Methot
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Zangwei Xu
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Haiyan Xu
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Dustin M. Smith
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Jennifer A. Piesvaux
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - J. Richard Miller
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Mark Bittinger
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Sheila H. Ranganath
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - David J. Bennett
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Erin F. DiMauro
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Alexander Pasternak
- Discovery Chemistry, Computational and Structural Chemistry, Quantitative Biosciences, Pharmacokinetics
and Drug Metabolism, Oncology Early Discovery, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| |
Collapse
|
30
|
Lau WL, Pearce B, Malakian H, Rodrigo I, Xie D, Gao M, Marsilio F, Chang C, Ruzanov M, Muckelbauer JK, Newitt JA, Lipovšek D, Sheriff S. Using yeast surface display to engineer a soluble and crystallizable construct of hematopoietic progenitor kinase 1 (HPK1). Acta Crystallogr F Struct Biol Commun 2021; 77:22-28. [PMID: 33439152 PMCID: PMC7805552 DOI: 10.1107/s2053230x20016015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/08/2020] [Indexed: 02/11/2023] Open
Abstract
Hematopoietic progenitor kinase 1 (HPK1) is an intracellular kinase that plays an important role in modulating tumor immune response and thus is an attractive target for drug discovery. Crystallization of the wild-type HPK1 kinase domain has been hampered by poor expression in recombinant systems and poor solubility. In this study, yeast surface display was applied to a library of HPK1 kinase-domain variants in order to select variants with an improved expression level and solubility. The HPK1 variant with the most improved properties contained two mutations, crystallized readily in complex with several small-molecule inhibitors and provided valuable insight to guide structure-based drug design. This work exemplifies the benefit of yeast surface display towards engineering crystallizable proteins and thus enabling structure-based drug discovery.
Collapse
Affiliation(s)
- Wai L. Lau
- Biologics Discovery, Bristol-Myers Squibb Research and Development, 100 Binney Street, Cambridge, MA 02142, USA
| | - Bradley Pearce
- Molecular Structure and Design, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Heather Malakian
- Biologics Discovery, Bristol-Myers Squibb Research and Development, 100 Binney Street, Cambridge, MA 02142, USA
| | - Iyoncy Rodrigo
- Protein Science, Bristol-Myers Squibb Research and Development, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Dianlin Xie
- Protein Science, Bristol-Myers Squibb Research and Development, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Mian Gao
- Protein Science, Bristol-Myers Squibb Research and Development, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Frank Marsilio
- Protein Science, Bristol-Myers Squibb Research and Development, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Chiehying Chang
- Molecular Structure and Design, Bristol-Myers Squibb Research and Development, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Max Ruzanov
- Molecular Structure and Design, Bristol-Myers Squibb Research and Development, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Jodi K. Muckelbauer
- Molecular Structure and Design, Bristol-Myers Squibb Research and Development, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - John A. Newitt
- Protein Science, Bristol-Myers Squibb Research and Development, PO Box 4000, Princeton, NJ 08543-4000, USA
| | - Daša Lipovšek
- Biologics Discovery, Bristol-Myers Squibb Research and Development, 100 Binney Street, Cambridge, MA 02142, USA
| | - Steven Sheriff
- Molecular Structure and Design, Bristol-Myers Squibb Research and Development, PO Box 4000, Princeton, NJ 08543-4000, USA
| |
Collapse
|
31
|
Wang Y, Zhang K, Georgiev P, Wells S, Xu H, Lacey BM, Xu Z, Laskey J, Mcleod R, Methot JL, Bittinger M, Pasternak A, Ranganath S. Pharmacological inhibition of hematopoietic progenitor kinase 1 positively regulates T-cell function. PLoS One 2020; 15:e0243145. [PMID: 33270695 PMCID: PMC7714195 DOI: 10.1371/journal.pone.0243145] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/17/2020] [Indexed: 12/18/2022] Open
Abstract
Hematopoietic progenitor kinase 1 (HPK1), a hematopoietic cell-specific Ste20-related serine/threonine kinase, is a negative regulator of signal transduction in immune cells, including T cells, B cells, and dendritic cells (DCs). In mice, HPK1 deficiency subverts inhibition of the anti-tumor immune response and is associated with functional augmentation of anti-tumor T cells. We have used a potent, small molecule HPK1 inhibitor, Compound 1, to investigate the effects of pharmacological intervention of HPK1 kinase activity in immune cells. Compound 1 enhanced Th1 cytokine production in T cells and fully reverted immune suppression imposed by the prostaglandin E2 (PGE2) and adenosine pathways in human T cells. Moreover, the combination of Compound 1 with pembrolizumab, a humanized monoclonal antibody against the programmed cell death protein 1 (PD-1), demonstrated a synergistic effect, resulting in enhanced interferon (IFN)-γ production. Collectively, our results suggest that blocking HPK1 kinase activity with small molecule inhibitors alone or in combination with checkpoint blockade may be an attractive approach for the immunotherapy of cancer.
Collapse
Affiliation(s)
- Yun Wang
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, United States of America
| | - Kelvin Zhang
- Department of Genetics and Pharmacogenomics, Merck & Co., Inc., Boston, Massachusetts, United States of America
| | - Peter Georgiev
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, United States of America
| | - Steven Wells
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, United States of America
| | - Haiyan Xu
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, Massachusetts, United States of America
| | - Brian M. Lacey
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, Massachusetts, United States of America
| | - Zangwei Xu
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, Massachusetts, United States of America
| | - Jason Laskey
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, Massachusetts, United States of America
| | - Robbie Mcleod
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, Massachusetts, United States of America
| | - Joey L. Methot
- Department of Discovery Chemistry, Merck & Co., Inc., Boston, Massachusetts, United States of America
| | - Mark Bittinger
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, United States of America
| | - Alexander Pasternak
- Department of Discovery Chemistry, Merck & Co., Inc., Boston, Massachusetts, United States of America
- * E-mail: (AP); (SR)
| | - Sheila Ranganath
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, United States of America
- * E-mail: (AP); (SR)
| |
Collapse
|
32
|
Activation of c-Jun N-Terminal Kinase, a Potential Therapeutic Target in Autoimmune Arthritis. Cells 2020; 9:cells9112466. [PMID: 33198301 PMCID: PMC7696795 DOI: 10.3390/cells9112466] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023] Open
Abstract
The c-Jun-N-terminal kinase (JNK) is a critical mediator involved in various physiological processes, such as immune responses, and the pathogenesis of various diseases, including autoimmune disorders. JNK is one of the crucial downstream signaling molecules of various immune triggers, mainly proinflammatory cytokines, in autoimmune arthritic conditions, mainly including rheumatoid arthritis, ankylosing spondylitis, and psoriatic arthritis. The activation of JNK is regulated in a complex manner by upstream kinases and phosphatases. Noticeably, different subtypes of JNKs behave differentially in immune responses. Furthermore, aside from biologics targeting proinflammatory cytokines, small-molecule inhibitors targeting signaling molecules such as Janus kinases can act as very powerful therapeutics in autoimmune arthritis patients unresponsiveness to conventional synthetic antirheumatic drugs. Nevertheless, despite these encouraging therapies, a population of patients with an inadequate therapeutic response to all currently available medications still remains. These findings identify the critical signaling molecule JNK as an attractive target for investigation of the immunopathogenesis of autoimmune disorders and for consideration as a potential therapeutic target for patients with autoimmune arthritis to achieve better disease control. This review provides a useful overview of the roles of JNK, how JNK is regulated in immunopathogenic responses, and the potential of therapeutically targeting JNK in patients with autoimmune arthritis.
Collapse
|
33
|
The functional analysis of Cullin 7 E3 ubiquitin ligases in cancer. Oncogenesis 2020; 9:98. [PMID: 33130829 PMCID: PMC7603503 DOI: 10.1038/s41389-020-00276-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 01/09/2023] Open
Abstract
Cullin (CUL) proteins have critical roles in development and cancer, however few studies on CUL7 have been reported due to its characteristic molecular structure. CUL7 forms a complex with the ROC1 ring finger protein, and only two F-box proteins Fbxw8 and Fbxw11 have been shown to bind to CUL7. Interestingly, CUL7 can interact with its substrates by forming a novel complex that is independent of these two F-box proteins. The biological implications of CUL-ring ligase 7 (CRL7) suggest that the CRL7 may not only perform a proteolytic function but may also play a non-proteolytic role. Among the existing studied CRL7-based E3 ligases, CUL7 exerts both tumor promotion and suppression in a context-dependent manner. Currently, the mechanism of CUL7 in cancer remains unclear, and no studies have addressed potential therapies targeting CUL7. Consistent with the roles of the various CRL7 adaptors exhibit, targeting CRL7 might be an effective strategy for cancer prevention and treatment. We systematically describe the recent major advances in understanding the role of the CUL7 E3 ligase in cancer and further summarize its potential use in clinical therapy.
Collapse
|
34
|
Sawasdikosol S, Burakoff S. A perspective on HPK1 as a novel immuno-oncology drug target. eLife 2020; 9:55122. [PMID: 32896273 PMCID: PMC7478889 DOI: 10.7554/elife.55122] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 08/28/2020] [Indexed: 12/11/2022] Open
Abstract
In this perspective review, the role Hematopoietic Progenitor Kinase 1 (HPK1) in tumor immunity will be reviewed, with special emphasis on how T cells are negatively-regulated at different junctures of cancer-immunity cycle by this regulatory kinase. The review will highlight the strengths and weaknesses of HPK1 as a candidate target for novel immuno-oncology (IO) drug development that is centered on the use of small molecule kinase inhibitor to modulate the immune response against cancer. Such a therapeutic approach, if proven successful, could supplement the cancer cell-centric standard of care therapies in order to fully meet the therapeutic needs of cancer patients.
Collapse
Affiliation(s)
- Sansana Sawasdikosol
- Tisch Cancer Institute, Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, Hess Center for Science and Medicine, New York, United States
| | - Steven Burakoff
- Tisch Cancer Institute, Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, Hess Center for Science and Medicine, New York, United States
| |
Collapse
|
35
|
Lacey BM, Xu Z, Chai X, Laskey J, Fradera X, Mittal P, Mishra S, Piesvaux J, Saradjian P, Shaffer L, Vassileva G, Gerdt C, Wang Y, Ferguson H, Smith DM, Ballard J, Wells S, Jain R, Mueller U, Addona G, Kariv I, Methot JL, Bittinger M, Ranganath S, Mcleod R, Pasternak A, Miller JR, Xu H. Development of High-Throughput Assays for Evaluation of Hematopoietic Progenitor Kinase 1 Inhibitors. SLAS DISCOVERY 2020; 26:88-99. [PMID: 32844715 DOI: 10.1177/2472555220952071] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Hematopoietic progenitor kinase 1 (HPK1), also referred to as mitogen-activated protein kinase kinase kinase kinase 1 (MAP4K1), is a serine/threonine kinase that negatively regulates T-cell signaling by phosphorylating Ser376 of Src homology 2 (SH2) domain-containing leukocyte protein of 76 kDa (SLP-76), a critical mediator of T-cell receptor activation. HPK1 loss of function mouse models demonstrated enhanced immune cell activation and beneficial antitumor activity. To enable discovery and functional characterization of high-affinity small-molecule HPK1 inhibitors, we have established high-throughput biochemical, cell-based, and novel pharmacodynamic (PD) assays. Kinase activity-based time-resolved fluorescence energy transfer (TR-FRET) assays were established as the primary biochemical approach to screen for potent inhibitors and assess selectivity against members of MAP4K and other closely related kinases. A proximal target engagement (TE) assay quantifying pSLP-76 levels as a readout and a distal assay measuring IL-2 secretion as a functional response were established using human peripheral blood mononuclear cells (PBMCs) from two healthy donors. Significant correlations between biochemical and cellular assays as well as excellent correlation between the two donors for the cellular assays were observed. pSLP-76 levels were further used as a PD marker in the preclinical murine model. This effort required the development of a novel ultrasensitive single-molecule array (SiMoA) assay to monitor pSLP-76 changes in mouse spleen.
Collapse
Affiliation(s)
- Brian M Lacey
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | - Zangwei Xu
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | - Xiaomei Chai
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | - Jason Laskey
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | - Xavier Fradera
- Department of Computational and Structural Chemistry, Merck & Co., Inc., Boston, MA, USA
| | - Payal Mittal
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, MA, USA
| | - Sasmita Mishra
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | - Jennifer Piesvaux
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | - Peter Saradjian
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | - Lynsey Shaffer
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | - Galya Vassileva
- Department of Genetics and Pharmacogenomics, Merck & Co., Inc., Boston, MA, USA
| | - Catherine Gerdt
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | - Yun Wang
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, MA, USA
| | - Heidi Ferguson
- Department of Preclinical Development, Merck & Co., Inc., Boston, MA, USA
| | | | | | - Steven Wells
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, MA, USA
| | - Rishabh Jain
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | - Uwe Mueller
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | - George Addona
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | - Ilona Kariv
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | - Joey L Methot
- Department of Discovery Chemistry, Merck & Co., Inc., Boston, MA, USA
| | - Mark Bittinger
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, MA, USA
| | - Sheila Ranganath
- Department of Oncology Early Discovery, Merck & Co., Inc., Boston, MA, USA
| | - Robbie Mcleod
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | | | - J Richard Miller
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | - Haiyan Xu
- Department of Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| |
Collapse
|
36
|
Song X, Akasaka H, Wang H, Abbasgholizadeh R, Shin JH, Zang F, Chen J, Logsdon CD, Maitra A, Bean AJ, Wang H. Hematopoietic progenitor kinase 1 down-regulates the oncogenic receptor tyrosine kinase AXL in pancreatic cancer. J Biol Chem 2020; 295:2348-2358. [PMID: 31959629 PMCID: PMC7039544 DOI: 10.1074/jbc.ra119.012186] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/14/2020] [Indexed: 12/23/2022] Open
Abstract
The oncogenic receptor tyrosine kinase AXL is overexpressed in cancer and plays an important role in carcinomas of multiple organs. However, the mechanisms of AXL overexpression in cancer remain unclear. In this study, using HEK293T, Panc-1, and Panc-28 cells and samples of human pancreatic intraepithelial neoplasia (PanIN), along with several biochemical approaches and immunofluorescence microscopy analyses, we sought to investigate the mechanisms that regulate AXL over-expression in pancreatic ductal adenocarcinoma (PDAC). We found that AXL interacts with hematopoietic progenitor kinase 1 (HPK1) and demonstrate that HPK1 down-regulates AXL and decreases its half-life. The HPK1-mediated AXL degradation was inhibited by the endocytic pathway inhibitors leupeptin, bafilomycin A1, and monensin. HPK1 accelerated the movement of AXL from the plasma membrane to endosomes in pancreatic cancer cells treated with the AXL ligand growth arrest-specific 6 (GAS6). Moreover, HPK1 increased the binding of AXL to the Cbl proto-oncogene (c-Cbl); promoted AXL ubiquitination; decreased AXL-mediated signaling, including phospho-AKT and phospho-ERK signaling; and decreased the invasion capability of PDAC cells. Importantly, we show that AXL expression inversely correlates with HPK1 expression in human PanINs and that patients whose tumors have low HPK1 and high AXL expression levels have shorter survival than those with low AXL or high HPK1 expression (p < 0.001). Our results suggest that HPK1 is a tumor suppressor that targets AXL for degradation via the endocytic pathway. HPK1 loss of function may contribute to AXL overexpression and thereby enhance AXL-dependent downstream signaling and tumor invasion in PDAC.
Collapse
Affiliation(s)
- Xianzhou Song
- Department of Anatomical Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Hironari Akasaka
- Department of Anatomical Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Hua Wang
- Department of Gastrointestinal Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Reza Abbasgholizadeh
- Department of Anatomical Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Ji-Hyun Shin
- Department of Anatomical Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Fenglin Zang
- Department of Anatomical Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Jiayi Chen
- Department of Anatomical Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Craig D Logsdon
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Anirban Maitra
- Department of Anatomical Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Andrew J Bean
- Department of Neurobiology and Anatomy, University of Texas McGovern Medical School, Houston, Texas 77030
| | - Huamin Wang
- Department of Anatomical Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030.
| |
Collapse
|
37
|
Chuang HC, Tan TH. MAP4K Family Kinases and DUSP Family Phosphatases in T-Cell Signaling and Systemic Lupus Erythematosus. Cells 2019; 8:cells8111433. [PMID: 31766293 PMCID: PMC6912701 DOI: 10.3390/cells8111433] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 12/21/2022] Open
Abstract
T cells play a critical role in the pathogenesis of systemic lupus erythematosus (SLE), which is a severe autoimmune disease. In the past 60 years, only one new therapeutic agent with limited efficacy has been approved for SLE treatment; therefore, the development of early diagnostic biomarkers and therapeutic targets for SLE is desirable. Mitogen-activated protein kinase kinase kinase kinases (MAP4Ks) and dual-specificity phosphatases (DUSPs) are regulators of MAP kinases. Several MAP4Ks and DUSPs are involved in T-cell signaling and autoimmune responses. HPK1 (MAP4K1), DUSP22 (JKAP), and DUSP14 are negative regulators of T-cell activation. Consistently, HPK1 and DUSP22 are downregulated in the T cells of human SLE patients. In contrast, MAP4K3 (GLK) is a positive regulator of T-cell signaling and T-cell-mediated immune responses. MAP4K3 overexpression-induced RORγt–AhR complex specifically controls interleukin 17A (IL-17A) production in T cells, leading to autoimmune responses. Consistently, MAP4K3 and the RORγt–AhR complex are overexpressed in the T cells of human SLE patients, as are DUSP4 and DUSP23. In addition, DUSPs are also involved in either human autoimmune diseases (DUSP2, DUSP7, DUSP10, and DUSP12) or T-cell activation (DUSP1, DUSP5, and DUSP14). In this review, we summarize the MAP4Ks and DUSPs that are potential biomarkers and/or therapeutic targets for SLE.
Collapse
|
38
|
Chen YH, Chen SH, Hou J, Ke ZB, Wu YP, Lin TT, Wei Y, Xue XY, Zheng QS, Huang JB, Xu N. Identifying hub genes of clear cell renal cell carcinoma associated with the proportion of regulatory T cells by weighted gene co-expression network analysis. Aging (Albany NY) 2019; 11:9478-9491. [PMID: 31672930 PMCID: PMC6874443 DOI: 10.18632/aging.102397] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/21/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Numerous patients with clear cell renal cell carcinoma (ccRCC) experience drug resistance after immunotherapy. Regulatory T (Treg) cells may work as a suppressor for anti-tumor immune response. PURPOSE We performed bioinformatics analysis to better understand the role of Treg cells in ccRCC. RESULTS Module 10 revealed the most relevance with Treg cells. Functional annotation showed that biological processes and pathways were mainly related to activation of the immune system and the processes of immunoreaction. Four hub genes were selected: LCK, MAP4K1, SLAMF6, and RHOH. Further validation showed that the four hub genes well-distinguished tumor and normal tissues and were good prognostic biomarkers for ccRCC. CONCLUSION The identified hub genes facilitate our knowledge of the underlying molecular mechanism of how Treg cells affect ccRCC in anti-tumor immune therapy. METHODS The CIBERSORT algorithm was performed to evaluate tumor-infiltrating immune cells based on the Cancer Genome Atlas cohort. Weighted gene co-expression network analysis was conducted to explore the modules related to Treg cells. Gene Ontology analysis and pathway enrichment analysis were performed for functional annotation and a protein-protein interaction network was built. Samples from the International Cancer Genomics Consortium database was used as a validation set.
Collapse
Affiliation(s)
- Ye-Hui Chen
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Shao-Hao Chen
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Jian Hou
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Zhi-Bin Ke
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Yu-Peng Wu
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Ting-Ting Lin
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Yong Wei
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Xue-Yi Xue
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Qing-Shui Zheng
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Jin-Bei Huang
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Ning Xu
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| |
Collapse
|
39
|
Chuang HC, Tan TH. MAP4K3/GLK in autoimmune disease, cancer and aging. J Biomed Sci 2019; 26:82. [PMID: 31640697 PMCID: PMC6806545 DOI: 10.1186/s12929-019-0570-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/01/2019] [Indexed: 01/01/2023] Open
Abstract
MAP4K3 (also named GLK) is a serine/threonine kinase, which belongs to the mammalian Ste20-like kinase family. At 22 years of age, GLK was initially cloned and identified as an upstream activator of the MAPK JNK under an environmental stress and proinflammatory cytokines. The data derived from GLK-overexpressing or shRNA-knockdown cell lines suggest that GLK may be involved in cell proliferation through mTOR signaling. GLK phosphorylates the transcription factor TFEB and retains TFEB in the cytoplasm, leading to inhibition of cell autophagy. After generating and characterizing GLK-deficient mice, the important in vivo roles of GLK in T-cell activation were revealed. In T cells, GLK directly interacts with and activates PKCθ through phosphorylating PKCθ at Ser-538 residue, leading to activation of IKK/NF-κB. Thus, GLK-deficient mice display impaired T-cell-mediated immune responses and decreased inflammatory phenotypes in autoimmune disease models. Consistently, the percentage of GLK-overexpressing T cells is increased in the peripheral blood from autoimmune disease patients; the GLK-overexpressing T cell population is correlated with disease severity of patients. The pathogenic mechanism of autoimmune disease by GLK overexpression was unraveled by characterizing T-cell-specific GLK transgenic mice and using biochemical analyses. GLK overexpression selectively promotes IL-17A transcription by inducing the AhR-RORγt complex in T cells. In addition, GLK overexpression in cancer tissues is correlated with cancer recurrence of human lung cancer and liver cancer; the predictive power of GLK overexpression for cancer recurrence is higher than that of pathologic stage. GLK directly phosphorylates and activates IQGAP1, resulting in induction of Cdc42-mediated cell migration and cancer metastasis. Furthermore, treatment of GLK inhibitor reduces disease severity of mouse autoimmune disease models and decreases IL-17A production of human autoimmune T cells. Due to the inhibitory function of HPK1/MAP4K1 in T-cell activation and the promoting effects of GLK on tumorigenesis, HPK1 and GLK dual inhibitors could be useful therapeutic drugs for cancer immunotherapy. In addition, GLK deficiency results in extension of lifespan in Caenorhabditis elegans and mice. Taken together, targeting MAP4K3 (GLK) may be useful for treating/preventing autoimmune disease, cancer metastasis/recurrence, and aging.
Collapse
Affiliation(s)
- Huai-Chia Chuang
- Immunology Research Center, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Taiwan
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Taiwan. .,Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
40
|
Wang Z, Han Z, Zhang L, Zhang S, Wang B. MicroRNA-98-5p regulates the proliferation and apoptosis of A549 cells by targeting MAP4K3. Oncol Lett 2019; 18:4288-4293. [PMID: 31579095 DOI: 10.3892/ol.2019.10771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/25/2019] [Indexed: 01/31/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is a primary subtype of lung cancer that is accompanied by a high incidence rate and poor prognosis. The primary treatment for NSCLC is chemotherapy, which has low effectiveness and high toxicity. Thus, novel targeted therapy has drawn much attention in recent years. MicroRNAs (miRs) serve important roles in multiple cancer types. In the current study, a decrease in miR-98-5p and an increase in mitogen-activated protein kinase kinase kinase kinase 3 (MAP4K3) was observed in NSCLC tumor tissues compared with normal tissues. miR-98-5p was predicted to target positions 1,056-1,063 of the MAP4K3 3'-untranslated region (UTR). The binding sites between miR-98-5p and the 3'-UTR of MAP4K3 messenger RNA were supported by the results of a dual-luciferase reporter assay. Compared with the control and miR-negative control (NC) groups, miR-98-5p mimic significantly reduced cell proliferation and increased apoptosis in NSCLC cells. In addition, miR-98-5p mimic reduced the expression of MAP4K3 and mammalian target of rapamycin while increasing the expression of cleaved caspase-3 compared with the control group and miR-NC groups. In conclusion, miR-98-5p may inhibit the progression of NSCLC via targeting of MAP4K3.
Collapse
Affiliation(s)
- Ziquan Wang
- Department of Medical Oncology, General Hospital of Xuzhou Mining Group, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China
| | - Zhengxiang Han
- Department of Medical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Lansheng Zhang
- Department of Medical Oncology, General Hospital of Xuzhou Mining Group, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China
| | - Shiqiang Zhang
- Department of Medical Oncology, General Hospital of Xuzhou Mining Group, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China
| | - Baoqing Wang
- Department of Medical Oncology, General Hospital of Xuzhou Mining Group, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China
| |
Collapse
|
41
|
Chuang HC, Chang CC, Teng CF, Hsueh CH, Chiu LL, Hsu PM, Lee MC, Hsu CP, Chen YR, Liu YC, Lyu PC, Tan TH. MAP4K3/GLK Promotes Lung Cancer Metastasis by Phosphorylating and Activating IQGAP1. Cancer Res 2019; 79:4978-4993. [DOI: 10.1158/0008-5472.can-19-1402] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/04/2019] [Accepted: 08/02/2019] [Indexed: 11/16/2022]
|
42
|
Batcha MM, Ahamed AS, Peng CF. Identification of a new type of haematopoietic progenitor kinase-interacting protein (HIP-55) in Aedes aegypti mosquito haemocytes and its involvement in immunity-like functions in mosquito: a molecular study. Parasitol Res 2019; 118:2509-2521. [PMID: 31377908 DOI: 10.1007/s00436-019-06408-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 07/25/2019] [Indexed: 11/26/2022]
Abstract
In this study, we characterize the HIP-55 protein in the mosquito Aedes aegypti for the first time. HIP-55 is a 55-kDa HPK1-interacting protein that is also called SH3P7. HIP-55 constitutively binds HPK1 'via' an HPK1 proline-rich motif 2(PR2) through its C-terminal SH3 domain. HIP-55 critically interacts with ZAP-70, and this interaction was induced by TCR signalling. ZAP-70 phosphorylated HIP-55 at Tyr-334 and Tyr-344 in vitro and in vivo. In our previous findings, AaZAP gene expression strongly proved that AaZAP-70 was involved in immunity-like functions in mosquito. Northern blot analysis of HIP-55 mRNA expression confirmed that it is only expressed in the abdomen and haemocyte tissues; this prediction correlates 100% and a polyclonal antibody also confirmed its localization in haemocytes and the abdomen. We prepared extracts to show the cytoplasmic expression (CE) of this protein. Previous results had proven that this protein is secreted from the cytoplasm; thus, we confirmed here that the protein is a cytoplasmic adaptor protein in mosquitoes and mammalian systems. Furthermore, our polyclonal antibody against HIP-55 also demonstrated that this protein is found in haemocytes and abdomen tissues, which assumes that the protein may be involved in phagocytic-like functions. RNAi (siRNA) silencing studies were used to degrade mosquito HIP-55; however, silencing only slightly affected the HIP-55 sequence and the gene transcriptional level. To characterize this protein, we cloned 609 bp from the 1.6-kb full-length cDNA using a pET28 vector for polyclonal antibody production. Graphical abstract.
Collapse
Affiliation(s)
- M Mohiadeen Batcha
- Department of Zoology, HKRH College, Uthamapalayam, Theni District, Tamil Nadu, India.
- Post Doctoral Scientist, Institute of Tropical Medicine, National Yang Ming University, Shihpai, Taipei-112, Taiwan, Republic of China.
| | - A Sajith Ahamed
- Department of Microbiology, HKRH College, Uthamapalayam, Theni District, Tamil Nadu, India
| | - Chiung Fang Peng
- Fuga Biotechnology, Chongqing S. Rd, Zhongzheng Dist, Taipei - 100, Taiwan
| |
Collapse
|
43
|
Liu J, Curtin J, You D, Hillerman S, Li-Wang B, Eraslan R, Xie J, Swanson J, Ho CP, Oppenheimer S, Warrack BM, McNaney CA, Nelson DM, Blum J, Kim T, Fereshteh M, Reily M, Shipkova P, Murtaza A, Sanjuan M, Hunt JT, Salter-Cid L. Critical role of kinase activity of hematopoietic progenitor kinase 1 in anti-tumor immune surveillance. PLoS One 2019; 14:e0212670. [PMID: 30913212 PMCID: PMC6435129 DOI: 10.1371/journal.pone.0212670] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 02/07/2019] [Indexed: 01/05/2023] Open
Abstract
Immunotherapy has fundamentally changed the landscape of cancer treatment. Despite the encouraging results with the checkpoint modulators, response rates vary widely across tumor types, with a majority of patients exhibiting either primary resistance without a significant initial response to treatment or acquired resistance with subsequent disease progression. Hematopoietic progenitor kinase 1 (HPK1) is predominantly expressed in hematopoietic cell linages and serves as a negative regulator in T cells and dendritic cells (DC). While HPK1 gene knockout (KO) studies suggest its role in anti-tumor immune responses, the involvement of kinase activity and thereof its therapeutic potential remain unknown. To investigate the potential of pharmacological intervention using inhibitors of HPK1, we generated HPK1 kinase dead (KD) mice which carry a single loss-of-function point mutation in the kinase domain and interrogated the role of kinase activity in immune cells in the context of suppressive factors or the tumor microenvironment (TME). Our data provide novel findings that HKP1 kinase activity is critical in conferring suppressive functions of HPK1 in a wide range of immune cells including CD4+, CD8+, DC, NK to Tregs, and inactivation of kinase domain was sufficient to elicit robust anti-tumor immune responses. These data support the concept that an HPK1 small molecule kinase inhibitor could serve as a novel agent to provide additional benefit in combination with existing immunotherapies, particularly to overcome resistance to current treatment regimens.
Collapse
Affiliation(s)
- Jinqi Liu
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Joshua Curtin
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Dan You
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Stephen Hillerman
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Bifang Li-Wang
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Rukiye Eraslan
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Jenny Xie
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Jesse Swanson
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Ching-Ping Ho
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Simone Oppenheimer
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Bethanne M. Warrack
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Colleen A. McNaney
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - David M. Nelson
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Jordan Blum
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Taeg Kim
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Mark Fereshteh
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Michael Reily
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Petia Shipkova
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Anwar Murtaza
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Miguel Sanjuan
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - John T. Hunt
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| | - Luisa Salter-Cid
- Immuno-oncology Discovery, Bristol Myers Squibb, Princeton, New Jersey, United States of America
| |
Collapse
|
44
|
Bai Y, Liu X, Qi X, Liu X, Peng F, Li H, Fu H, Pei S, Chen L, Chi X, Zhang L, Zhu X, Song Y, Wang Y, Meng S, Jiang T, Shao S. PDIA6 modulates apoptosis and autophagy of non-small cell lung cancer cells via the MAP4K1/JNK signaling pathway. EBioMedicine 2019; 42:311-325. [PMID: 30922965 PMCID: PMC6491656 DOI: 10.1016/j.ebiom.2019.03.045] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/15/2019] [Accepted: 03/15/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the most common type of lung cancer with a poor prognosis. We previously found that protein disulfide isomerase family 6 (PDIA6) is upregulated in lung squamous cell carcinoma (LSCC). This study aimed to elucidate the clinical relevance, biological functions, and molecular mechanisms of PDIA6 in NSCLC. METHODS The expression of PDIA6 in NSCLC was assessed using the TCGA database, western blotting, and immunohistochemistry. Correlations of PDIA6 expression with clinicopathological and survival features were evaluated. The functions of PDIA6 in regulating NSCLC cell growth, apoptosis, and autophagy were investigated using gain-and loss-of-function strategies in vitro or in vivo. The underlying molecular mechanisms of PDIA6 function were examined by human phospho-kinase array and co-immunoprecipitation. FINDINGS PDIA6 expression was upregulated in NSCLC compared with adjacent normal tissues, and the higher PDIA6 expression was correlated with poor prognosis. PDIA6 knockdown decreased NSCLC cell proliferation and increased cisplatin-induced intrinsic apoptosis, while PDIA6 overexpression had the opposite effects. In addition, PDIA6 regulated cisplatin-induced autophagy, and this contributed to PDIA6-mediated apoptosis in NSCLC cells. Mechanistically, PDIA6 reduced the phosphorylation levels of JNK and c-Jun. Moreover, PDIA6 interacted with MAP4K1 and inhibited its phosphorylation, ultimately inhibiting the JNK/c-Jun signaling pathway. INTERPRETATION PDIA6 is overexpressed in NSCLC and inhibits cisplatin-induced NSCLC cell apoptosis and autophagy via the MAP4K1/JNK/c-Jun signaling pathway, suggesting that PDIA6 may serve as a biomarker and therapeutic target for NSCLC patients. FUND: National Natural Science Foundation of China and Institutions of higher learning of innovation team from Liaoning province.
Collapse
Affiliation(s)
- Yuxin Bai
- Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Xuefeng Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Xiaoyu Qi
- Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Xuan Liu
- Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Fang Peng
- Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Huimin Li
- Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Hailu Fu
- Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Shimei Pei
- Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Liying Chen
- Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Xinming Chi
- Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Liyuan Zhang
- Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Xinbing Zhu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Yang Song
- Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Yang Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Songshu Meng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Tao Jiang
- Department of Andrology, The First Hospital Affiliated of Dalian Medical University, Dalian 116011, China.
| | - Shujuan Shao
- Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
45
|
Wu P, Sneeringer CJ, Pitts KE, Day ES, Chan BK, Wei B, Lehoux I, Mortara K, Li H, Wu J, Franke Y, Moffat JG, Grogan JL, Heffron TP, Wang W. Hematopoietic Progenitor Kinase-1 Structure in a Domain-Swapped Dimer. Structure 2018; 27:125-133.e4. [PMID: 30503777 DOI: 10.1016/j.str.2018.10.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 07/27/2018] [Accepted: 10/25/2018] [Indexed: 12/12/2022]
Abstract
Enhancement of antigen-specific T cell immunity has shown significant therapeutic benefit in infectious diseases and cancer. Hematopoietic progenitor kinase-1 (HPK1) is a negative-feedback regulator of T cell receptor signaling, which dampens T cell proliferation and effector function. A recent report showed that a catalytic dead mutant of HPK1 phenocopies augmented T cell responses observed in HPK1-knockout mice, indicating that kinase activity is critical for function. We evaluated active and inactive mutants and determined crystal structures of HPK1 kinase domain (HPK1-KD) in apo and ligand bound forms. In all structures HPK1-KD displays a rare domain-swapped dimer, in which the activation segment comprises a well-conserved dimer interface. Biophysical measurements show formation of dimer in solution. The activation segment adopts an α-helical structure which exhibits distinct orientations in active and inactive states. This face-to-face configuration suggests that the domain-swapped dimer may possess alternative selectivity for certain substrates of HPK1 under relevant cellular context.
Collapse
Affiliation(s)
- Ping Wu
- Department of Structural Biology, Genentech, South San Francisco, CA 94080, USA
| | | | - Keith E Pitts
- Department of Biochemical Pharmacology, Genentech, South San Francisco, CA 94080, USA
| | - Eric S Day
- Department of Late Stage Pharmaceutical Development, Genentech, South San Francisco, CA 94080, USA
| | - Bryan K Chan
- Department of Discovery Chemistry, Genentech, South San Francisco, CA 94080, USA
| | - Binqing Wei
- Department of Discovery Chemistry, Genentech, South San Francisco, CA 94080, USA
| | - Isabelle Lehoux
- Department of Biomolecular Resources, Genentech, South San Francisco, CA 94080, USA
| | - Kyle Mortara
- Department of Biomolecular Resources, Genentech, South San Francisco, CA 94080, USA
| | - Hong Li
- Department of Protein Chemistry, Genentech, South San Francisco, CA 94080, USA
| | - Jiansheng Wu
- Department of Protein Chemistry, Genentech, South San Francisco, CA 94080, USA
| | - Yvonne Franke
- Department of Biomolecular Resources, Genentech, South San Francisco, CA 94080, USA
| | - John G Moffat
- Department of Biochemical Pharmacology, Genentech, South San Francisco, CA 94080, USA
| | - Jane L Grogan
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Timothy P Heffron
- Department of Discovery Chemistry, Genentech, South San Francisco, CA 94080, USA
| | - Weiru Wang
- Department of Structural Biology, Genentech, South San Francisco, CA 94080, USA.
| |
Collapse
|
46
|
Hernandez S, Qing J, Thibodeau RH, Du X, Park S, Lee HM, Xu M, Oh S, Navarro A, Roose-Girma M, Newman RJ, Warming S, Nannini M, Sampath D, Kim JM, Grogan JL, Mellman I. The Kinase Activity of Hematopoietic Progenitor Kinase 1 Is Essential for the Regulation of T Cell Function. Cell Rep 2018; 25:80-94. [DOI: 10.1016/j.celrep.2018.09.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/12/2018] [Accepted: 09/07/2018] [Indexed: 01/20/2023] Open
|
47
|
Hsu CP, Chuang HC, Lee MC, Tsou HH, Lee LW, Li JP, Tan TH. GLK/MAP4K3 overexpression associates with recurrence risk for non-small cell lung cancer. Oncotarget 2018; 7:41748-41757. [PMID: 27203390 PMCID: PMC5173093 DOI: 10.18632/oncotarget.9410] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/23/2016] [Indexed: 01/14/2023] Open
Abstract
Lung cancer is the leading cause of cancer death worldwide. Non-small cell lung cancer (NSCLC) accounts for 85% of total lung cancers; 40% to 60% of NSCLC patients die of cancer recurrence after cancer resection. Since GLK (also named MAP4K3) induces activation of NF-κB, which contributes to tumor progression, we investigated the role of GLK in NSCLC. GLK protein levels of 190 samples from pulmonary tissue arrays and 58 pulmonary resection samples from stage I to stage III NSCLC patients were studied using immunohistochemistry or immunoblotting. High levels of GLK proteins were detected in pulmonary tissues from NSCLC patients. Elevated GLK protein levels were correlated with increased recurrence risks and poor recurrence-free survival rates in NSCLC patients after adjusting for pathologic stage, smoking status, alcohol status, and EGFR levels. Thus, GLK is a novel prognostic biomarker for NSCLC recurrence.
Collapse
Affiliation(s)
- Chung-Ping Hsu
- Division of Thoracic Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung, 40705, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Huai-Chia Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Ming-Ching Lee
- Division of Thoracic Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung, 40705, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Hsiao-Hui Tsou
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Li-Wen Lee
- Division of Thoracic Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung, 40705, Taiwan
| | - Ju-Pi Li
- Immunology Research Center, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan, 35053, Taiwan.,Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, 77030, USA
| |
Collapse
|
48
|
Zeng L, Wang WH, Arrington J, Shao G, Geahlen RL, Hu CD, Tao WA. Identification of Upstream Kinases by Fluorescence Complementation Mass Spectrometry. ACS CENTRAL SCIENCE 2017; 3:1078-1085. [PMID: 29104924 PMCID: PMC5658758 DOI: 10.1021/acscentsci.7b00261] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Indexed: 05/09/2023]
Abstract
Protein kinases and their substrates comprise extensive signaling networks that regulate many diverse cellular functions. However, methods and techniques to systematically identify kinases directly responsible for specific phosphorylation events have remained elusive. Here we describe a novel proteomic strategy termed fluorescence complementation mass spectrometry (FCMS) to identify kinase-substrate pairs in high throughput. The FCMS strategy employs a specific substrate and a kinase library, both of which are fused with fluorescence complemented protein fragments. Transient and weak kinase-substrate interactions in living cells are stabilized by the association of fluorescence protein fragments. These kinase-substrate pairs are then isolated with high specificity and are identified and quantified by LC-MS. FCMS was applied to the identification of both known and novel kinases of the transcription factor, cAMP response element-binding protein (CREB). Novel CREB kinases were validated by in vitro kinase assays, and the phosphorylation sites were unambiguously located. These results uncovered possible new roles for CREB in multiple important signaling pathways and demonstrated the great potential of this new proteomic strategy.
Collapse
Affiliation(s)
- Lingfei Zeng
- Department
of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Wen-Horng Wang
- Department
of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Justine Arrington
- Department
of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Gengbao Shao
- Department
of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Robert L. Geahlen
- Department
of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue
Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, United States
| | - Chang-Deng Hu
- Department
of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue
Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, United States
| | - W. Andy Tao
- Department
of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
- Department
of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Department
of Biochemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue
Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, United States
- E-mail:
| |
Collapse
|
49
|
Zhang Q, Ding S, Zhang H. Interactions between hematopoietic progenitor kinase 1 and its adaptor proteins. Mol Med Rep 2017; 16:6472-6482. [DOI: 10.3892/mmr.2017.7494] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 07/31/2017] [Indexed: 11/06/2022] Open
|
50
|
Chuang HC, Tan TH. MAP4K4 and IL-6 + Th17 cells play important roles in non-obese type 2 diabetes. J Biomed Sci 2017; 24:4. [PMID: 28061846 PMCID: PMC5219747 DOI: 10.1186/s12929-016-0307-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 12/05/2016] [Indexed: 12/24/2022] Open
Abstract
Obesity is a causal factor of type 2 diabetes (T2D); however, people without obesity (including lean, normal weight, or overweight) may still develop T2D. Non-obese T2D is prevalent in Asia and also frequently occurs in Europe. Recently, multiple evidences oppose the notion that either obesity or central obesity (visceral fat accumulation) promotes non-obese T2D. Several factors such as inflammation and environmental factors contribute to non-obese T2D. According to the data derived from gene knockout mice and T2D clinical samples in Asia and Europe, the pathogenesis of non-obese T2D has been unveiled recently. MAP4K4 downregulation in T cells results in enhancement of the IL-6+ Th17 cell population, leading to insulin resistance and T2D in both human and mice. Moreover, MAP4K4 single nucleotide polymorphisms and epigenetic changes are associated with T2D patients. Interactions between MAP4K4 gene variants and environmental factors may contribute to MAP4K4 attenuation in T cells, leading to non-obese T2D. Future investigations of the pathogenesis of non-obese T2D shall lead to development of precision medicine for non-obese T2D.
Collapse
Affiliation(s)
- Huai-Chia Chuang
- Immunology Research Center, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Taiwan
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Taiwan. .,Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, 77030, USA.
| |
Collapse
|