1
|
Ando Y, Horiuchi Y, Hatazawa S, Mataki M, Nakamura A, Murakami T. Hyperdifferentiated murine melanoma cells promote adaptive anti-tumor immunity but activate the immune checkpoint system. Oncoimmunology 2025; 14:2437211. [PMID: 39648330 PMCID: PMC11633153 DOI: 10.1080/2162402x.2024.2437211] [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: 04/28/2024] [Revised: 11/06/2024] [Accepted: 11/28/2024] [Indexed: 12/10/2024] Open
Abstract
Accumulating evidence suggests that phenotype switching of cancer cells is essential for therapeutic resistance. However, the immunological characteristics of drug-induced phenotype-switching melanoma cells (PSMCs) are unknown. We investigated PSMC elimination by host immunity using hyperdifferentiated melanoma model cells derived from murine B16F10 melanoma cells. Exposure of B16F10 cells to staurosporine induced a hyperdifferentiated phenotype associated with transient drug tolerance. Staurosporine-induced hyperdifferentiated B16F10 (sB16F10) cells expressed calreticulin on their surface and were phagocytosed efficiently. Furthermore, the inoculation of mice with sB16F10 cells induced immune responses against tumor-derived antigens. Despite the immunogenicity of sB16F10 cells, they activated the PD-1/PD-L1 immune checkpoint system and strongly resisted T cell-mediated tumor destruction. However, in vivo treatment with immune checkpoint inhibitors successfully eliminated the tumor. Thus, hyperdifferentiated melanoma cells have conflicting immunological properties - enhanced immunogenicity and immune evasion. Inhibiting the ability of PSMCs to evade T cell-mediated elimination might lead to complete melanoma eradication.
Collapse
Affiliation(s)
- Yukie Ando
- Department of Microbiology, Saitama Medical University, Moroyama-cho, Saitama,Japan
| | - Yutaka Horiuchi
- Department of Microbiology, Saitama Medical University, Moroyama-cho, Saitama,Japan
| | - Sara Hatazawa
- Department of Microbiology, Saitama Medical University, Moroyama-cho, Saitama,Japan
| | - Momo Mataki
- Department of Microbiology, Saitama Medical University, Moroyama-cho, Saitama,Japan
| | - Akihiro Nakamura
- Department of Microbiology, Saitama Medical University, Moroyama-cho, Saitama,Japan
| | - Takashi Murakami
- Department of Microbiology, Saitama Medical University, Moroyama-cho, Saitama,Japan
| |
Collapse
|
2
|
Isinelli G, Failla S, Plebani R, Prete A. Exploring oncology treatment strategies with tyrosine kinase inhibitors through advanced 3D models (Review). MEDICINE INTERNATIONAL 2025; 5:13. [PMID: 39790707 PMCID: PMC11707505 DOI: 10.3892/mi.2024.212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025]
Abstract
The limitations of two-dimensional (2D) models in cancer research have hindered progress in fully understanding the complexities of drug resistance and therapeutic failures. However, three-dimensional (3D) models provide a more accurate representation of in vivo environments, capturing critical cellular interactions and dynamics that are essential in evaluating the efficacy and toxicity of tyrosine kinase inhibitors (TKIs). These advanced models enable researchers to explore drug resistance mechanisms with greater precision, optimizing treatment strategies and improving the predictive accuracy of clinical outcomes. By leveraging 3D models, it will be possible to deepen the current understanding of TKIs and drive forward innovations in cancer treatment. The present review discusses the limitations of 2D models and the transformative impact of 3D models on oncology research, highlighting their roles in addressing the challenges of 2D systems and advancing TKI studies.
Collapse
Affiliation(s)
- Giorgia Isinelli
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Chemistry, Biology and Biotechnology, University of Perugia, I-06123 Perugia, Italy
| | - Sharon Failla
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Roberto Plebani
- Department of Medical, Oral and Biotechnological Sciences, ‘G. D'Annunzio’ University, I-66100 Chieti-Pescara, Italy
| | - Alessandro Prete
- Department of Clinical and Experimental Medicine, Endocrine Unit 2, University of Pisa, I-56122 Pisa, Italy
| |
Collapse
|
3
|
Jassim BA, Bai Y, Qu Z, Sander CJ, Lin J, Miao J, Zhang ZY. Structure-activity relationship studies and design of a PTPN22 inhibitor with enhanced isozyme selectivity and cellular efficacy. Eur J Med Chem 2025; 283:117129. [PMID: 39693863 PMCID: PMC11709133 DOI: 10.1016/j.ejmech.2024.117129] [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: 10/15/2024] [Revised: 11/28/2024] [Accepted: 11/30/2024] [Indexed: 12/20/2024]
Abstract
Protein tyrosine phosphatase non-receptor type 22 (PTPN22) lies downstream of the T cell receptor (TCR) and attenuates T cell signaling by dephosphorylating key effector proteins such as LCK, Zap70, and the intracellular region of the TCR. Recent evidence implicates PTPN22 as an exciting target for enabling immunotherapeutic efficacy against cancer. We carried out structural optimization of a benzofuran salicylic acid-based orthosteric PTPN22 inhibitor 8b, using a combination of crystal structure analysis, synthesis, matched molecular pairs analysis, and biochemical and cell-based assays. Herein, we report structure-activity relationship studies, lead optimization based on the 8b-PTPN22 co-crystal structure, and cellular evaluation of the top analog. Notably, our efforts yielded compound 8b-19, an essentially equipotent scaffold with superior isozyme selectivity, improved aqueous solubility, and significantly enhanced cellular efficacy compared to the parent 8b. This compound may serve as a lead for further optimization of PTPN22-targeting immunotherapies or as a chemical probe for interrogation for additional links between PTPN22 and immunomodulation in cells.
Collapse
Affiliation(s)
- Brenson A Jassim
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Yunpeng Bai
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Zihan Qu
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Conrad J Sander
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Jianping Lin
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Jinmin Miao
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Zhong-Yin Zhang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA; Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA; Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
4
|
Xu G, Zhang Q, Cheng R, Qu J, Li W. Survival strategies of cancer cells: the role of macropinocytosis in nutrient acquisition, metabolic reprogramming, and therapeutic targeting. Autophagy 2025. [PMID: 39817564 DOI: 10.1080/15548627.2025.2452149] [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: 10/09/2024] [Revised: 12/27/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025] Open
Abstract
Macropinocytosis is a nonselective form of endocytosis that allows cancer cells to largely take up the extracellular fluid and its contents, including nutrients, growth factors, etc. We first elaborate meticulously on the process of macropinocytosis. Only by thoroughly understanding this entire process can we devise targeted strategies against it. We then focus on the central role of the MTOR (mechanistic target of rapamycin kinase) complex 1 (MTORC1) in regulating macropinocytosis, highlighting its significance as a key signaling hub where various pathways converge to control nutrient uptake and metabolic processes. The article covers a comprehensive analysis of the literature on the molecular mechanisms governing macropinocytosis, including the initiation, maturation, and recycling of macropinosomes, with an emphasis on how these processes are hijacked by cancer cells to sustain their growth. Key discussions include the potential therapeutic strategies targeting macropinocytosis, such as enhancing drug delivery via this pathway, inhibiting macropinocytosis to starve cancer cells, blocking the degradation and recycling of macropinosomes, and inducing methuosis - a form of cell death triggered by excessive macropinocytosis. Targeting macropinocytosis represents a novel and innovative approach that could significantly advance the treatment of cancers that rely on this pathway for survival. Through continuous research and innovation, we look forward to developing more effective and safer anti-cancer therapies that will bring new hope to patients.
Collapse
Affiliation(s)
- Guoshuai Xu
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| | - Qinghong Zhang
- Emergency Department, Shengjing Hospital of China Medical University, Liaoning, China
| | - Renjia Cheng
- Department of Intensive Care Medicine, The General Hospital of the Northern Theater Command of the People's Liberation Army of China, Liaoning, China
| | - Jun Qu
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| | - Wenqiang Li
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| |
Collapse
|
5
|
Ghosh AK, Chen Y, Gadi RK, Sonawane A, Gamage SP, Tesmer JG. Design, synthesis, and X-ray structural studies of a series of highly potent, selective, and drug-like G protein-coupled receptor kinase 5 inhibitors. Eur J Med Chem 2025; 282:117024. [PMID: 39549325 DOI: 10.1016/j.ejmech.2024.117024] [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: 08/16/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024]
Abstract
G protein-coupled receptor kinase 5 (GRK5) has emerged as a potential drug development target against heart failure and cancer. A close homolog, GRK6 represents a therapeutic target for multiple myeloma. We have rationally designed a series of highly selective, potent, noncovalent, and drug-like GRK5 inhibitors. Several inhibitors exhibited low nanomolar GRK5 inhibition and high selectivity over GRK2, and, surprisingly, some were selective for GRK6. We determined high-resolution X-ray crystal structures of several inhibitors in complex with GRK5, which provide molecular insights into the ligand-binding site interactions responsible for GRK5 selectivity and potency.
Collapse
Affiliation(s)
- Arun K Ghosh
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA; Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA.
| | - Yueyi Chen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Ranjith Kumar Gadi
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Amol Sonawane
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Sandali Piladuwa Gamage
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - JohnJ G Tesmer
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
6
|
Shi Z, Artemenko M, Yu W, Zhang M, Yi C, Chen P, Lin S, Bian Z, Lian B, Meng F, Chen J, Roussel T, Li Y, Chan KKL, Ip PPC, Lai HC, To SKY, Liu X, Peng L, Wong AST. Bola-Amphiphilic Dendrimer Enhances Imatinib to Target Metastatic Ovarian Cancer via β-Catenin-HRP2 Signaling Axis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:2884-2898. [PMID: 39752231 DOI: 10.1021/acsami.4c12857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Ovarian cancer is the leading cause of death among all gynecological malignancies, and drug resistance renders the current chemotherapy agents ineffective for patients with advanced metastatic tumors. We report an effective treatment strategy for targeting metastatic ovarian cancer involving a nanoformulation (Bola/IM)─bola-amphiphilic dendrimer (Bola)-encapsulated imatinib (IM)─to target the critical mediator of ovarian cancer stem cells (CSCs) CD117 (c-Kit). Bola/IM offered significantly more effective targeting of CSCs compared to IM alone, through a novel and tumor-specific β-catenin/HRP2 axis, allowing potent inhibition of cancer cell survival, stemness, and metastasis in metastatic and drug-resistant ovarian cancer cells. Promising results were also obtained in clinically relevant patient-derived ascites and organoids alongside high tumor-oriented accumulation and favorable pharmacokinetic properties in mouse models. Furthermore, Bola/IM displayed synergistic anticancer activity when combined with the first-line chemotherapeutic drug cisplatin in patient-derived xenograft mouse models without any adverse effects. Our findings support the use of Bola/IM as a nanoformulation to empower IM, providing targeted and potent treatment of metastatic ovarian cancer. Our study thus represents a significant advancement toward addressing the unmet medical need for improved therapies targeting this challenging disease.
Collapse
Affiliation(s)
- Zeyu Shi
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong 999077, China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Pokfulam, Hong Kong 999077, China
| | - Margarita Artemenko
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong 999077, China
| | - Weiyu Yu
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong 999077, China
| | - Ming Zhang
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong 999077, China
| | - Canhui Yi
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong 999077, China
| | - Peng Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Shuting Lin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Zhancun Bian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
- Aix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, Equipe Labellisée Ligue Contre le Cancer, 13288 Marseille, France
| | - Baoping Lian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Fanzhen Meng
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Jiaxuan Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
- Aix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, Equipe Labellisée Ligue Contre le Cancer, 13288 Marseille, France
| | - Tom Roussel
- Aix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, Equipe Labellisée Ligue Contre le Cancer, 13288 Marseille, France
| | - Ying Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Karen K L Chan
- Department of Obstetrics and Gynecology, Queen Mary Hospital, University of Hong Kong, Pokfulam, Hong Kong 999077, China
| | - Philip P C Ip
- Department of Pathology, Queen Mary Hospital, University of Hong Kong, Pokfulam, Hong Kong 999077, China
| | - Hung-Cheng Lai
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, Taipei 23561, Taiwan
| | - Sally K Y To
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong 999077, China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Pokfulam, Hong Kong 999077, China
| | - Xiaoxuan Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Ling Peng
- Aix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, Equipe Labellisée Ligue Contre le Cancer, 13288 Marseille, France
| | - Alice S T Wong
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong 999077, China
| |
Collapse
|
7
|
Lee J, Bang D, Kim S. Residue-Level Multiview Deep Learning for ATP Binding Site Prediction and Applications in Kinase Inhibitors. J Chem Inf Model 2025; 65:50-61. [PMID: 39690486 DOI: 10.1021/acs.jcim.4c01255] [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: 12/19/2024]
Abstract
Accurate identification of adenosine triphosphate (ATP) binding sites is crucial for understanding cellular functions and advancing drug discovery, particularly in targeting kinases for cancer treatment. Existing methods face significant challenges due to their reliance on time-consuming precomputed features and the heavily imbalanced nature of binding site data without further investigations on their utility in drug discovery. To address these limitations, we introduced Multiview-ATPBind and ResiBoost. Multiview-ATPBind is an end-to-end deep learning model that integrates one-dimensional (1D) sequence and three-dimensional (3D) structural information for rapid and precise residue-level pocket-ligand interaction predictions. Additionally, ResiBoost is a novel residue-level boosting algorithm designed to mitigate data imbalance by enhancing the prediction of rare positive binding residues. Our approach outperforms state-of-the-art models on benchmark data sets, showing significant improvements in balanced metrics with both experimental and AI-predicted structures. Furthermore, our model seamlessly transfers to predicting binding sites and enhancing docking simulations for kinase inhibitors, including imatinib and dasatinib, underscoring the potential of our method in drug discovery applications.
Collapse
Affiliation(s)
- Jaechan Lee
- Department of Computer Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
- AIGENDRUG Co., Ltd., Seoul 08826, Republic of Korea
| | - Dongmin Bang
- AIGENDRUG Co., Ltd., Seoul 08826, Republic of Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Republic of Korea
| | - Sun Kim
- Department of Computer Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
- AIGENDRUG Co., Ltd., Seoul 08826, Republic of Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Republic of Korea
- Interdisciplinary Program in Artificial Intelligence, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
8
|
Lutomski CA, Bennett JL, El-Baba TJ, Wu D, Hinkle JD, Burnap SA, Liko I, Mullen C, Syka JEP, Struwe WB, Robinson CV. Defining proteoform-specific interactions for drug targeting in a native cell signalling environment. Nat Chem 2025:10.1038/s41557-024-01711-w. [PMID: 39806141 DOI: 10.1038/s41557-024-01711-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025]
Abstract
Understanding the dynamics of membrane protein-ligand interactions within a native lipid bilayer is a major goal for drug discovery. Typically, cell-based assays are used, however, they are often blind to the effects of protein modifications. In this study, using the archetypal G protein-coupled receptor rhodopsin, we found that the receptor and its effectors can be released directly from retina rod disc membranes using infrared irradiation in a mass spectrometer. Subsequent isolation and dissociation by infrared multiphoton dissociation enabled the sequencing of individual retina proteoforms. Specifically, we categorized distinct proteoforms of rhodopsin, localized labile palmitoylations, discovered a Gβγ proteoform that abolishes membrane association and defined lipid modifications on G proteins that influence their assembly. Given reports of undesirable side-effects involving vision, we characterized the off-target drug binding of two phosphodiesterase 5 inhibitors, vardenafil and sildenafil, to the retina rod phosphodiesterase 6 (PDE6). The results demonstrate differential off-target reactivity with PDE6 and an interaction preference for lipidated proteoforms of G proteins. In summary, this study highlights the opportunities for probing proteoform-ligand interactions within natural membrane environments.
Collapse
Affiliation(s)
- Corinne A Lutomski
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Jack L Bennett
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Tarick J El-Baba
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Di Wu
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | | | - Sean A Burnap
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
| | | | | | | | - Weston B Struwe
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Carol V Robinson
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK.
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK.
| |
Collapse
|
9
|
Lin TE, Chou CH, Wu YW, Sung TY, Hsu JY, Yen SC, Hsieh JH, Chang YW, Pan SL, Huang WJ, Hsu KC, Yang CR. Identification of a Potent CDK8 Inhibitor Using Structure-Based Virtual Screening. J Chem Inf Model 2025; 65:378-389. [PMID: 39740163 PMCID: PMC11733953 DOI: 10.1021/acs.jcim.4c02011] [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: 10/30/2024] [Revised: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025]
Abstract
Pulmonary fibrosis is excessive scarring of the lung tissues. Transforming growth factor-beta (TGF-β) has been implicated in pulmonary fibrosis due to its ability to induce the epithelial-to-mesenchymal transition (EMT) and promote epithelial cell migration. Cyclin-dependent kinase 8 (CDK8) can mediate the TGF-β signaling pathways and could function as an alternative therapeutic target for treating pulmonary fibrosis. Here, we performed a structure-based virtual screening campaign to identify CDK8 inhibitors from a library of 1.6 million compounds. The screening process ended with the identification of a novel CDK8 inhibitor, P162-0948 (IC50: 50.4 nM). An interaction analysis highlighted important CDK8-ligand interactions that support its binding and inhibitory activity. Testing against a panel of 60 different kinases demonstrated P162-0948 selectivity toward CDK8. Crucially, the inhibitor was found to be structurally novel when compared to known CDK8 inhibitors. Testing in A549 human alveolar epithelial cell lines showed that the P162-0948 can reduce cell migration and protein expression of EMT-related proteins. When P162-0948 was treated in cells at 5 μM, phosphorylation of Smad in the nucleus was reduced, which suggests disruption of the TGF-β/Smad signaling pathway. The identification of P162-0948 shows that it is not only potent, but its structural novelty can inform future design studies for potential therapeutics targeting pulmonary fibrosis.
Collapse
Affiliation(s)
- Tony Eight Lin
- Graduate
Institute of Cancer Biology and Drug Discovery, College of Medical
Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Ph.D. Program
for Cancer Molecular Biology and Drug Discovery, College of Medical
Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Ching-Hsuan Chou
- School of
Pharmacy, College of Medicine, National
Taiwan University, Taipei 10051, Taiwan
| | - Yi-Wen Wu
- Graduate
Institute of Cancer Biology and Drug Discovery, College of Medical
Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Tzu-Ying Sung
- Graduate
Institute of Cancer Biology and Drug Discovery, College of Medical
Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Jui-Yi Hsu
- Graduate
Institute of Cancer Biology and Drug Discovery, College of Medical
Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Ph.D. Program
for Cancer Molecular Biology and Drug Discovery, College of Medical
Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Shih-Chung Yen
- Warshel
Institute
for Computational Biology, The Chinese University
of Hong Kong (Shenzhen), Shenzhen, Guangdong 518172, People’s Republic of China
| | - Jui-Hua Hsieh
- Division
of Translational Toxicology, National Institute of Environmental Health
Sciences, National Institutes of Health, Durham, North Carolina 27709-2233, United
States
| | - Yu-Wei Chang
- Ph.D. Program
for Cancer Molecular Biology and Drug Discovery, College of Medical
Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Department
of Traditional Chinese Medicine, Chang Gung
Memorial Hospital, Keelung Medical Center, Keelung 20401,Taiwan
| | - Shiow-Lin Pan
- Graduate
Institute of Cancer Biology and Drug Discovery, College of Medical
Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Ph.D. Program
for Cancer Molecular Biology and Drug Discovery, College of Medical
Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Ph.D. Program
in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research
Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031,Taiwan
| | - Wei-Jan Huang
- Ph.D. Program
in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
- School of
Pharmacy, Taipei Medical University, Taipei 10051, Taiwan
| | - Kai-Cheng Hsu
- Graduate
Institute of Cancer Biology and Drug Discovery, College of Medical
Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Ph.D. Program
for Cancer Molecular Biology and Drug Discovery, College of Medical
Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Ph.D. Program
in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research
Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031,Taiwan
- Cancer
Center, Wan Fang Hospital, Taipei Medical
University, Taipei 11696,Taiwan
| | - Chia-Ron Yang
- School of
Pharmacy, College of Medicine, National
Taiwan University, Taipei 10051, Taiwan
| |
Collapse
|
10
|
Kim JE, Ko W, Jin S, Woo JN, Jung Y, Bae I, Choe HK, Seo D, Hille B, Suh BC. Activation of TMEM16E scramblase induces ligand independent growth factor receptor signaling and macropinocytosis for membrane repair. Commun Biol 2025; 8:35. [PMID: 39794444 PMCID: PMC11724107 DOI: 10.1038/s42003-025-07465-6] [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: 03/28/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
The calcium-dependent phospholipid scramblase TMEM16E mediates ion transport and lipid translocation across the plasma membrane. TMEM16E also contributes to protection of membrane structure by facilitating cellular repair signaling. Our research reveals that TMEM16E activation promotes macropinocytosis, essential for maintaining plasma membrane integrity. This scramblase externalizes phosphatidylserine, typically linked to resting growth factor receptors. We demonstrate that TMEM16E can interact with and signal through growth factor receptors, including epidermal growth factor receptor, even without ligands. This interaction stimulates downstream phosphoinositide 3-kinase and facilitates macropinocytosis and internalization of annexin V bound to the membrane, a process sensitive to amiloride inhibition. Although TMEM16E is internalized during this process, it returns to the plasma membrane. TMEM16E- driven macropinocytosis is proposed to restore membrane integrity after perturbation, potentially explaining pathologies in conditions like muscular dystrophies, where TMEM16E functionality is compromised, highlighting its critical role in muscle cell survival.
Collapse
Affiliation(s)
- Jung-Eun Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Woori Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Siwoo Jin
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Jin-Nyeong Woo
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Yuna Jung
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Inah Bae
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Han-Kyoung Choe
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Daeha Seo
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Bertil Hille
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Byung-Chang Suh
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea.
| |
Collapse
|
11
|
Wen X, McLaren DG. High-throughput hit identification with acoustic ejection mass spectrometry. SLAS Technol 2025; 31:100245. [PMID: 39800101 DOI: 10.1016/j.slast.2025.100245] [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: 11/05/2024] [Revised: 12/17/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
This mini-review provides an overview of recent developments in AEMS supporting hit identification in drug discovery, emphasizing its potential to enhance the quality and efficiency of label-free HTS. Future advancements that may further expand the role of AEMS in the drug discovery process will also be discussed.
Collapse
|
12
|
Haag A, Němec V, Janovská P, Bartošíková J, Adhikari B, Müller J, Schwalm MP, Čada Š, Ohmayer U, Daub H, Kim Y, Born F, Wolf E, Bryja V, Knapp S. Development and Discovery of a Selective Degrader of Casein Kinases 1 δ/ε. J Med Chem 2025; 68:506-530. [PMID: 39729064 DOI: 10.1021/acs.jmedchem.4c02201] [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: 12/28/2024]
Abstract
Members of the casein kinase 1 (CK1) family have emerged as key regulators of cellular signaling and as potential drug targets. Functional annotation of the 7 human isoforms would benefit from isoform-selective inhibitors, allowing studies on the role of these enzymes in normal physiology and disease pathogenesis. However, due to significant sequence homology within the catalytic domain, isoform selectivity is difficult to achieve with conventional small molecules. Here, we used a PROTAC (Proteolysis TArgeting Chimeras) approach to develop a highly selective degrader AH078 (37) targeting CK1δ and CK1ε with excellent selectivity over the highly related CK1α isoform. The developed PROTAC, AH078 (37) selectively degraded CK1δ and CK1ε with a DC50 of 200 nM. Characterization of AH078 (37) revealed a VHL and Ubiquitin-dependent degradation mechanism. Thus, AH078 (37) represents a versatile chemical tool to study CK1δ and CK1ε function in cellular systems.
Collapse
Affiliation(s)
- Adrian Haag
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Václav Němec
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Pavlína Janovská
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Jana Bartošíková
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Bikash Adhikari
- Institute of Biochemistry, University of Kiel, Rudolf-Höber-Str. 1, Kiel 24118, Germany
| | - Juliane Müller
- Institute of Biochemistry, University of Kiel, Rudolf-Höber-Str. 1, Kiel 24118, Germany
| | - Martin P Schwalm
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Štěpán Čada
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Uli Ohmayer
- NEOsphere Biotechnologies GmbH, Fraunhoferstr. 1, 82152 Martinsried, Germany
| | - Henrik Daub
- NEOsphere Biotechnologies GmbH, Fraunhoferstr. 1, 82152 Martinsried, Germany
| | - Yeojin Kim
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Florian Born
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Elmar Wolf
- Institute of Biochemistry, University of Kiel, Rudolf-Höber-Str. 1, Kiel 24118, Germany
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
- German translational cancer network (DKTK) site Frankfurt Mainz, 60590 Heidelberg, Germany
| |
Collapse
|
13
|
Wu J, Liu Y, Zhang Y, Wang X, Yan H, Zhu Y, Song J, Yu DJ. Identifying Protein-Nucleotide Binding Residues via Grouped Multi-task Learning and Pre-trained Protein Language Models. J Chem Inf Model 2025. [PMID: 39788787 DOI: 10.1021/acs.jcim.4c02092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
The accurate identification of protein-nucleotide binding residues is crucial for protein function annotation and drug discovery. Numerous computational methods have been proposed to predict these binding residues, achieving remarkable performance. However, due to the limited availability and high variability of nucleotides, predicting binding residues for diverse nucleotides remains a significant challenge. To address these, we propose NucGMTL, a new grouped deep multi-task learning approach designed for predicting binding residues of all observed nucleotides in the BioLiP database. NucGMTL leverages pre-trained protein language models to generate robust sequence embedding and incorporates multi-scale learning along with scale-based self-attention mechanisms to capture a broader range of feature dependencies. To effectively harness the shared binding patterns across various nucleotides, deep multi-task learning is utilized to distill common representations, taking advantage of auxiliary information from similar nucleotides selected based on task grouping. Performance evaluation on benchmark data sets shows that NucGMTL achieves an average area under the Precision-Recall curve (AUPRC) of 0.594, surpassing other state-of-the-art methods. Further analyses highlight that the predominant advantage of NucGMTL can be reflected by its effective integration of grouped multi-task learning and pre-trained protein language models. The data set and source code are freely accessible at: https://github.com/jerry1984Y/NucGMTL.
Collapse
Affiliation(s)
- Jiashun Wu
- School of Computer Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Yan Liu
- School of Information Engineering, Yangzhou University, Yangzhou 225100, China
| | - Ying Zhang
- School of Computer Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Xiaoyu Wang
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - He Yan
- College of Information Science and Technology & Artificial Intelligence, Nanjing Forestry University, Nanjing 210037, China
| | - Yiheng Zhu
- College of Artificial Intelligence, Nanjing Agricultural University, Nanjing 210095, China
| | - Jiangning Song
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
- Monash Data Futures Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Dong-Jun Yu
- School of Computer Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| |
Collapse
|
14
|
Sander P, Schwalm MP, Krämer A, Elson L, Rasch A, Masberg B, Selig R, Sievers-Engler A, Lämmerhofer M, Müller S, Knapp S, Albrecht W, Laufer SA. Design, Synthesis, and Biochemical Evaluation of Novel MLK3 Inhibitors: A Target Hopping Example. J Med Chem 2025; 68:674-694. [PMID: 39681301 DOI: 10.1021/acs.jmedchem.4c02552] [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: 12/18/2024]
Abstract
The human kinome has tremendous medical potential. In the past decade, mixed-lineage protein kinase 3 (MLK3) has emerged as an interesting and druggable target in oncogenic signaling. The important role of MLK3 has been demonstrated in several types of cancer. In a target hopping example we started with the focal adhesion kinase (FAK) inhibitor PF-431396 (10), which shows off-target activity toward MLK3. We were able to develop highly active compounds in the single digit nanomolar range for MLK3. Furthermore, we achieved a dramatic shift in selectivity from FAK to MLK3. Here we present a new chemical class of MLK3 inhibitors, including our lead compound 37 with an outstanding IC50 value of <1 nM in a biochemical MLK3 assay while simultaneously exhibiting kinome-wide selectivity.
Collapse
Affiliation(s)
- Pascal Sander
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tuebingen, Auf der Morgenstelle 8, Tuebingen 72076, Germany
| | - Martin P Schwalm
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), DTKT Site Frankfurt-Mainz, Heidelberg 69120, Germany
| | - Andreas Krämer
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Lewis Elson
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Alexander Rasch
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tuebingen, Auf der Morgenstelle 8, Tuebingen 72076, Germany
| | - Benedikt Masberg
- Pharmaceutical (Bio-)Analysis, Institute of Pharmaceutical Sciences, Eberhard-Karls University of Tuebingen, Auf der Morgenstelle 8, Tuebingen 72076, Germany
| | - Roland Selig
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tuebingen, Auf der Morgenstelle 8, Tuebingen 72076, Germany
- HepaRegeniX GmbH, Eisenbahnstraße 63, Tuebingen 72072, Germany
| | - Adrian Sievers-Engler
- Pharmaceutical (Bio-)Analysis, Institute of Pharmaceutical Sciences, Eberhard-Karls University of Tuebingen, Auf der Morgenstelle 8, Tuebingen 72076, Germany
| | - Michael Lämmerhofer
- Pharmaceutical (Bio-)Analysis, Institute of Pharmaceutical Sciences, Eberhard-Karls University of Tuebingen, Auf der Morgenstelle 8, Tuebingen 72076, Germany
| | - Susanne Müller
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, Frankfurt am Main 60438, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, Frankfurt am Main 60438, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), DTKT Site Frankfurt-Mainz, Heidelberg 69120, Germany
| | | | - Stefan A Laufer
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tuebingen, Auf der Morgenstelle 8, Tuebingen 72076, Germany
- Cluster of Excellence "Image Guided and Functionally Instructed Tumor Therapies" (iFIT), Eberhard Karls University of Tuebingen, Tuebingen 72076, Germany
- German Consortium for Translational Cancer Research (DKTK), Partner Site Tuebingen, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
- Tuebingen Center for Academic Drug Discovery, Auf der Morgenstelle 8, Tuebingen 72076, Germany
| |
Collapse
|
15
|
Müller J, Bayer FP, Wilhelm M, Schuh MG, Kuster B, The M. PTMNavigator: interactive visualization of differentially regulated post-translational modifications in cellular signaling pathways. Nat Commun 2025; 16:510. [PMID: 39779715 PMCID: PMC11711753 DOI: 10.1038/s41467-024-55533-y] [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: 11/28/2023] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Post-translational modifications (PTMs) play pivotal roles in regulating cellular signaling, fine-tuning protein function, and orchestrating complex biological processes. Despite their importance, the lack of comprehensive tools for studying PTMs from a pathway-centric perspective has limited our ability to understand how PTMs modulate cellular pathways on a molecular level. Here, we present PTMNavigator, a tool integrated into the ProteomicsDB platform that offers an interactive interface for researchers to overlay experimental PTM data with pathway diagrams. PTMNavigator provides ~3000 canonical pathways from manually curated databases, enabling users to modify and create custom diagrams tailored to their data. Additionally, PTMNavigator automatically runs kinase and pathway enrichment algorithms whose results are directly integrated into the visualization. This offers a comprehensive view of the intricate relationship between PTMs and signaling pathways. We demonstrate the utility of PTMNavigator by applying it to two phosphoproteomics datasets, showing how it can enhance pathway enrichment analysis, visualize how drug treatments result in a discernable flow of PTM-driven signaling, and aid in proposing extensions to existing pathways. By enhancing our understanding of cellular signaling dynamics and facilitating the discovery of PTM-pathway interactions, PTMNavigator advances our knowledge of PTM biology and its implications in health and disease.
Collapse
Affiliation(s)
- Julian Müller
- Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Florian P Bayer
- Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Mathias Wilhelm
- Computational Mass Spectrometry, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Maximilian G Schuh
- Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
- Organic Chemistry II, School of Natural Sciences, Technical University of Munich, Garching, Germany
| | - Bernhard Kuster
- Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Matthew The
- Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany.
| |
Collapse
|
16
|
Stadnicki EJ, Ludewig H, Kumar RP, Wang X, Qiao Y, Kern D, Bradshaw N. Dual-action kinase inhibitors influence p38α MAP kinase dephosphorylation. Proc Natl Acad Sci U S A 2025; 122:e2415150122. [PMID: 39739785 PMCID: PMC11725910 DOI: 10.1073/pnas.2415150122] [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: 08/09/2024] [Accepted: 12/03/2024] [Indexed: 01/02/2025] Open
Abstract
Reversible protein phosphorylation directs essential cellular processes including cell division, cell growth, cell death, inflammation, and differentiation. Because protein phosphorylation drives diverse diseases, kinases and phosphatases have been targets for drug discovery, with some achieving remarkable clinical success. Most protein kinases are activated by phosphorylation of their activation loops, which shifts the conformational equilibrium of the kinase toward the active state. To turn off the kinase, protein phosphatases dephosphorylate these sites, but how the conformation of the dynamic activation loop contributes to dephosphorylation was not known. To answer this, we modulated the activation loop conformational equilibrium of human p38α ΜΑP kinase with existing kinase inhibitors that bind and stabilize specific inactive activation loop conformations. From this, we identified three inhibitors that increase the rate of dephosphorylation of the activation loop phospho-threonine by the PPM serine/threonine phosphatase WIP1. Hence, these compounds are "dual-action" inhibitors that simultaneously block the active site and promote p38α dephosphorylation. Our X-ray crystal structures of phosphorylated p38α bound to the dual-action inhibitors reveal a shared flipped conformation of the activation loop with a fully accessible phospho-threonine. In contrast, our X-ray crystal structure of phosphorylated apo human p38α reveals a different activation loop conformation with an inaccessible phospho-threonine, thereby explaining the increased rate of dephosphorylation upon inhibitor binding. These findings reveal a conformational preference of phosphatases for their targets and suggest a unique approach to achieving improved potency and specificity for therapeutic kinase inhibitors.
Collapse
Affiliation(s)
| | - Hannes Ludewig
- Department of Biochemistry, Brandeis University, Waltham, MA02454
- HHMI, Brandeis University, Waltham, MA02454
| | | | - Xicong Wang
- Department of Biochemistry, Brandeis University, Waltham, MA02454
| | - Youwei Qiao
- Department of Biochemistry, Brandeis University, Waltham, MA02454
| | - Dorothee Kern
- Department of Biochemistry, Brandeis University, Waltham, MA02454
- HHMI, Brandeis University, Waltham, MA02454
| | - Niels Bradshaw
- Department of Biochemistry, Brandeis University, Waltham, MA02454
| |
Collapse
|
17
|
Moyano PM, Kubina T, Paruch ŠO, Jarošková A, Novotný J, Skočková V, Ovesná P, Suchánková T, Prokofeva P, Kuster B, Šmída M, Chaikuad A, Krämer A, Knapp S, Souček K, Paruch K. Thieno[3,2-b]pyridine: Attractive scaffold for highly selective inhibitors of underexplored protein kinases with variable binding mode. Angew Chem Int Ed Engl 2025; 64:e202412786. [PMID: 39503260 DOI: 10.1002/anie.202412786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Indexed: 11/22/2024]
Abstract
Protein kinases are key regulators of numerous biological processes and aberrant kinase activity can cause various diseases, particularly cancer. Herein, we report the identification of new series of highly selective kinase inhibitors based on the thieno[3,2-b]pyridine scaffold. The weak interaction of the thieno[3,2-b]pyridine core with the kinase hinge region allows for profoundly different binding modes all of which maintain high kinome-wide selectivity, as illustrated by the isomers MU1464 and MU1668. Thus, this core structure provides a template of ATP-competitive but not ATP-mimetic inhibitors that are anchored at the kinase back pocket. Mapping the chemical space around the central thieno[3,2-b]pyridine pharmacophore afforded highly selective inhibitors of the kinase Haspin, exemplified by the compound MU1920 that fulfils criteria for a quality chemical probe and is suitable for use in in vivo applications. However, despite the role of Haspin in mitosis, the inhibition of Haspin alone was not sufficient to elicit cytotoxic effect in cancer cells. The thieno[3,2-b]pyridine scaffold can be used in a broader context, as a basis of inhibitors targeting other underexplored protein kinases, such as CDKLs.
Collapse
Affiliation(s)
- Paula Martín Moyano
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
- International Clinical Research Centre, St. Anne's University Hospital, Pekařská 53, Brno, 656 91, Czech Republic
| | - Tadeáš Kubina
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Štěpán Owen Paruch
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Aneta Jarošková
- Institute of Biophysics, Czech Academy of Science, Královopolská 135, 612 00, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Jan Novotný
- Institute of Biophysics, Czech Academy of Science, Královopolská 135, 612 00, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Veronika Skočková
- Institute of Biophysics, Czech Academy of Science, Královopolská 135, 612 00, Brno, Czech Republic
| | - Petra Ovesná
- Institute of Biostatistics and Analyses, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Tereza Suchánková
- Institute of Biophysics, Czech Academy of Science, Královopolská 135, 612 00, Brno, Czech Republic
| | - Polina Prokofeva
- Proteomics and Bioanalytics, Department of Molecular Life Sciences, School of Life Sciences, Technical University of Munich, Freising, 85354, Germany
| | - Bernhard Kuster
- Proteomics and Bioanalytics, Department of Molecular Life Sciences, School of Life Sciences, Technical University of Munich, Freising, 85354, Germany
| | - Michal Šmída
- CEITEC, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Apirat Chaikuad
- Institute for Pharmaceutical Chemistry, Structural Genomics Consortium, Johann Wolfgang Goethe-University, Max-von-Laue-Strasse 15, Frankfurt am Main, 60438, Germany
| | - Andreas Krämer
- Institute for Pharmaceutical Chemistry, Structural Genomics Consortium, Johann Wolfgang Goethe-University, Max-von-Laue-Strasse 15, Frankfurt am Main, 60438, Germany
| | - Stefan Knapp
- Institute for Pharmaceutical Chemistry, Structural Genomics Consortium, Johann Wolfgang Goethe-University, Max-von-Laue-Strasse 15, Frankfurt am Main, 60438, Germany
| | - Karel Souček
- International Clinical Research Centre, St. Anne's University Hospital, Pekařská 53, Brno, 656 91, Czech Republic
- Institute of Biophysics, Czech Academy of Science, Královopolská 135, 612 00, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Kamil Paruch
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
- International Clinical Research Centre, St. Anne's University Hospital, Pekařská 53, Brno, 656 91, Czech Republic
| |
Collapse
|
18
|
Wu D, Sun Q, Tang H, Xiao H, Luo J, Ouyang L, Sun Q. Acquired resistance to tyrosine kinase targeted therapy: mechanism and tackling strategies. Drug Resist Updat 2025; 78:101176. [PMID: 39642660 DOI: 10.1016/j.drup.2024.101176] [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: 09/17/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 12/09/2024]
Abstract
Over the past two decades, tyrosine kinase inhibitors (TKIs) have rapidly emerged as pivotal targeted agents, offering promising therapeutic prospects for patients. However, as the cornerstone of targeted therapies, an increasing number of TKIs have been found to develop acquired resistance during treatment, making the challenge of overcoming this resistance a primary focus of current research. This review comprehensively examines the evolution of TKIs from multiple perspectives, with particular emphasis on the mechanisms underlying acquired resistance, innovative drug design strategies, inherent challenges, and future directions.
Collapse
Affiliation(s)
- Defa Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Qian Sun
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China; West China Medical Publishers, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haolin Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Huan Xiao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Jiaxiang Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China.
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China; West China Medical Publishers, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
19
|
Cao Y, Yu T, Zhu Z, Zhang Y, Sun S, Li N, Gu C, Yang Y. Exploring the landscape of post-translational modification in drug discovery. Pharmacol Ther 2025; 265:108749. [PMID: 39557344 DOI: 10.1016/j.pharmthera.2024.108749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/11/2024] [Accepted: 11/04/2024] [Indexed: 11/20/2024]
Abstract
Post-translational modifications (PTMs) play a crucial role in regulating protein function, and their dysregulation is frequently associated with various diseases. The emergence of epigenetic drugs targeting factors such as histone deacetylases (HDACs) and histone methyltransferase enhancers of zeste homolog 2 (EZH2) has led to a significant shift towards precision medicine, offering new possibilities to overcome the limitations of traditional therapeutics. In this review, we aim to systematically explore how small molecules modulate PTMs. We discuss the direct targeting of enzymes involved in PTM pathways, the modulation of substrate proteins, and the disruption of protein-enzyme interactions that govern PTM processes. Additionally, we delve into the emerging strategy of employing multifunctional molecules to precisely regulate the modification levels of proteins of interest (POIs). Furthermore, we examine the specific characteristics of these molecules, evaluating their therapeutic benefits and potential drawbacks. The goal of this review is to provide a comprehensive understanding of PTM-targeting strategies and their potential for personalized medicine, offering a forward-looking perspective on the evolution of precision therapeutics.
Collapse
Affiliation(s)
- Yuhao Cao
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tianyi Yu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ziang Zhu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuanjiao Zhang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shanliang Sun
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Nianguang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Chunyan Gu
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Ye Yang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
20
|
Joseph RE, Wales TE, Jayne S, Britton RG, Fulton DB, Engen JR, Dyer MJS, Andreotti AH. Impact of the clinically approved BTK inhibitors on the conformation of full-length BTK and analysis of the development of BTK resistance mutations in chronic lymphocytic leukemia. eLife 2024; 13:RP95488. [PMID: 39728925 DOI: 10.7554/elife.95488] [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] [Indexed: 12/28/2024] Open
Abstract
Inhibition of Bruton's tyrosine kinase (BTK) has proven to be highly effective in the treatment of B-cell malignancies such as chronic lymphocytic leukemia (CLL), autoimmune disorders, and multiple sclerosis. Since the approval of the first BTK inhibitor (BTKi), Ibrutinib, several other inhibitors including Acalabrutinib, Zanubrutinib, Tirabrutinib, and Pirtobrutinib have been clinically approved. All are covalent active site inhibitors, with the exception of the reversible active site inhibitor Pirtobrutinib. The large number of available inhibitors for the BTK target creates challenges in choosing the most appropriate BTKi for treatment. Side-by-side comparisons in CLL have shown that different inhibitors may differ in their treatment efficacy. Moreover, the nature of the resistance mutations that arise in patients appears to depend on the specific BTKi administered. We have previously shown that Ibrutinib binding to the kinase active site causes unanticipated long-range effects on the global conformation of BTK (Joseph et al., 2020). Here, we show that binding of each of the five approved BTKi to the kinase active site brings about distinct allosteric changes that alter the conformational equilibrium of full-length BTK. Additionally, we provide an explanation for the resistance mutation bias observed in CLL patients treated with different BTKi and characterize the mechanism of action of two common resistance mutations: BTK T474I and L528W.
Collapse
Affiliation(s)
- Raji E Joseph
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, United States
| | - Thomas E Wales
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, United States
| | - Sandrine Jayne
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, College of Life Sciences, University of Leicester, Leicester, United Kingdom
| | - Robert G Britton
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, College of Life Sciences, University of Leicester, Leicester, United Kingdom
| | - D Bruce Fulton
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, United States
| | - John R Engen
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, United States
| | - Martin J S Dyer
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, College of Life Sciences, University of Leicester, Leicester, United Kingdom
| | - Amy H Andreotti
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, United States
| |
Collapse
|
21
|
Zhang Q, Yu J, You Q, Wang L. Modulating Phosphorylation by Proximity-Inducing Modalities for Cancer Therapy. J Med Chem 2024; 67:21695-21716. [PMID: 39648992 DOI: 10.1021/acs.jmedchem.4c02624] [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: 12/10/2024]
Abstract
Abnormal phosphorylation of proteins can lead to various diseases, particularly cancer. Therefore, the development of small molecules for precise regulation of protein phosphorylation holds great potential for drug design. While the traditional kinase/phosphatase small-molecule modulators have shown some success, achieving precise phosphorylation regulation has proven to be challenging. The emergence of heterobifunctional molecules, such as phosphorylation-inducing chimeric small molecules (PHICSs) and phosphatase recruiting chimeras (PHORCs), with proximity-inducing modalities is expected to lead to a breakthrough by specifically recruiting kinase or phosphatase to the protein of interest. Herein, we summarize the drug targets with aberrant phosphorylation in cancer and underscore the potential of correcting phosphorylation in cancer therapy. Through reported cases of heterobifunctional molecules targeting phosphorylation regulation, we highlight the current design strategies and features of these molecules. We also provide a systematic elaboration of the link between aberrantly phosphorylated targets and cancer as well as the existing challenges and future research directions for developing heterobifunctional molecular drugs for phosphorylation regulation.
Collapse
Affiliation(s)
- Qiuyue Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jia Yu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
22
|
Wang G, Seidler NJ, Röhm S, Pan Y, Liang XJ, Haarer L, Berger BT, Sivashanmugam SA, Wydra VR, Forster M, Laufer SA, Chaikuad A, Gehringer M, Knapp S. Probing the Protein Kinases' Cysteinome by Covalent Fragments. Angew Chem Int Ed Engl 2024:e202419736. [PMID: 39716901 DOI: 10.1002/anie.202419736] [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: 10/12/2024] [Revised: 11/23/2024] [Accepted: 12/09/2024] [Indexed: 12/25/2024]
Abstract
Protein kinases are important drug targets, yet specific inhibitors have been developed for only a fraction of the more than 500 human kinases. A major challenge in designing inhibitors for highly related kinases is selectivity. Unlike their non-covalent counterparts, covalent inhibitors offer the advantage of selectively targeting structurally similar kinases by modifying specific protein side chains, particularly non-conserved cysteines. Previously, covalent fragment screens yielded potent and selective inhibitors for individual kinases such as ERK1/2 but have not been applied to the broader kinome. Furthermore, many of the accessible cysteine positions have not been addressed so far. Here, we outline a generalizable approach to sample ATP-site cysteines with fragment-like covalent inhibitors. We present the development of a kinase-focused covalent fragment library and its systematic screening against a curated selection of 47 kinases, with 60 active site-proximal cysteines using LC/MS and differential scanning fluorimetry (DSF) assays, followed by hit validation through various complementary techniques. Our findings expand the repertoire of targetable cysteines within protein kinases, provide insight into unique binding modes identified from crystal structures and deliver isoform-specific hits with promising profiles as starting points for the development of highly potent and selective covalent inhibitors.
Collapse
Affiliation(s)
- Guiqun Wang
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), DKTK Site Frankfurt-Mainz, German Cancer Research Center (DKFZ), D-69120, Heidelberg, Germany
| | - Nico J Seidler
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
| | - Sandra Röhm
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Yufeng Pan
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Xiaojun Julia Liang
- Faculty of Medicine, Institute for Biomedical Engineering, Department for Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Lisa Haarer
- Faculty of Medicine, Institute for Biomedical Engineering, Department for Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Benedict-Tilman Berger
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Saran Aswathaman Sivashanmugam
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Valentin R Wydra
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
| | - Michael Forster
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Stefan A Laufer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- Tübingen Center for Academic Drug Discovery & Development (TüCAD2), Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Apirat Chaikuad
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Matthias Gehringer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Faculty of Medicine, Institute for Biomedical Engineering, Department for Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Stefan Knapp
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), DKTK Site Frankfurt-Mainz, German Cancer Research Center (DKFZ), D-69120, Heidelberg, Germany
| |
Collapse
|
23
|
Zhao Z, Bourne PE. Exploring Extended Warheads toward Developing Cysteine-Targeted Covalent Kinase Inhibitors. J Chem Inf Model 2024; 64:9517-9527. [PMID: 39656065 DOI: 10.1021/acs.jcim.4c00890] [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: 12/24/2024]
Abstract
In designing covalent kinase inhibitors (CKIs), the inclusion of electrophiles as attacking warheads demands careful choreography, ensuring not only their presence on the scaffold moiety but also their precise interaction with nucleophiles in the binding sites. Given the limited number of known electrophiles, exploring adjacent chemical space to broaden the palette of available electrophiles capable of covalent inhibition is desirable. Here, we systematically analyze the characteristics of warheads and the corresponding adjacent fragments for use in CKI design. We first collect all the released cysteine-targeted CKIs from multiple databases and create one CKI data set containing 16,961 kinase-inhibitor data points from 12,381 unique CKIs covering 146 kinases with accessible cysteines in their binding pockets. Then, we analyze this data set, focusing on the extended warheads (i.e., warheads + adjacent fragments)─including 30 common warheads and 1344 unique adjacent fragments. In so doing, we provide structural insights and delineate chemical properties and patterns in these extended warheads. Notably, we highlight the popular patterns observed within reversible CKIs for the popular warheads cyanoacrylamide and aldehyde. This study provides medicinal chemists with novel insights into extended warheads and a comprehensive source of adjacent fragments, thus guiding the design, synthesis, and optimization of CKIs.
Collapse
Affiliation(s)
- Zheng Zhao
- School of Data Science and Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Philip E Bourne
- School of Data Science and Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22904, United States
| |
Collapse
|
24
|
Goodis CC, Eberly C, Chan AM, Kim M, Lowe BD, Civin CI, Fletcher S. The polypharmacy combination of the BCL-2 inhibitor venetoclax (VEN) and the FLT3 inhibitor gilteritinib (GIL) is more active in acute myeloid leukemia cells than novel polypharmacologic BCL-2/FLT3 VEN-GIL hybrid single-molecule inhibitors. Eur J Med Chem 2024; 285:117190. [PMID: 39813774 DOI: 10.1016/j.ejmech.2024.117190] [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: 10/28/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/18/2025]
Abstract
Current treatments for acute myeloid leukemias (AMLs) cure fewer than 30 % of patients. This low efficacy is due, in part, to the inter-patient and intra-patient heterogeneity of AMLs; accordingly, all current AML treatment regimens involve drug combinations (polypharmacy). A recently-completed clinical trial in relapsed/refractory AML using a combination of two newer targeted antileukemics, the BCL-2 inhibitor venetoclax (VEN) plus the FLT3 inhibitor gilteritinib (GIL), yielded highly promising results for this two-drug polypharmacy combination. Polypharmacology - wherein a single drug molecule that inhibits two or more biological targets is created - has been proposed to offer superior therapeutic results, as compared to the corresponding polypharmacy approach. Herein, we designed and synthesized several polypharmacologic dual BCL-2/FLT3 hybrid single-molecule inhibitors by tethering VEN to GIL, through their solvent-exposed domains. While the in vitro antileukemic activity of the two-drug VEN + GIL polypharmacy combination proved superior to our focused library of VEN-GIL hybrids, alternative grafting points on GIL may yield improved results for future hybrid compounds.
Collapse
Affiliation(s)
- Christopher C Goodis
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine St., Baltimore, MD, 21201, USA
| | - Christian Eberly
- Center for Stem Cell Biology & Regenerative Medicine, Department of Pediatrics, University of Maryland School of Medicine, 20 Penn St., Room S103, Baltimore, MD, 21201, USA
| | - Alexandria M Chan
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine St., Baltimore, MD, 21201, USA
| | - MinJung Kim
- Center for Stem Cell Biology & Regenerative Medicine, Department of Pediatrics, University of Maryland School of Medicine, 20 Penn St., Room S103, Baltimore, MD, 21201, USA
| | - Brandon D Lowe
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine St., Baltimore, MD, 21201, USA
| | - Curt I Civin
- Center for Stem Cell Biology & Regenerative Medicine, Departments of Pediatrics and Pharmacology, Physiology & Drug Development, University of Maryland School of Medicine, 20 Penn St., Room S103, Baltimore, MD, 21201, USA; University of Maryland Marlene & Stewart Greenebaum Comprehensive Cancer Center, 22 S. Greene St., Baltimore, MD, 21201, USA
| | - Steven Fletcher
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine St., Baltimore, MD, 21201, USA; University of Maryland Marlene & Stewart Greenebaum Comprehensive Cancer Center, 22 S. Greene St., Baltimore, MD, 21201, USA.
| |
Collapse
|
25
|
Al Mazid MF, Shkel O, Ryu E, Kim J, Shin KH, Kim YK, Lim HS, Lee JS. Aptamer and N-Degron Ensemble (AptaGron) as a Target Protein Degradation Strategy. ACS Chem Biol 2024; 19:2462-2468. [PMID: 39630150 DOI: 10.1021/acschembio.4c00536] [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: 12/21/2024]
Abstract
Target protein degradation (TPD) is a promising strategy for catalytic downregulation of target proteins through various cellular proteolytic pathways. Despite numerous reports on novel TPD mechanisms, the discovery of target-specific ligands remains a major challenge. Unlike small-molecule ligands, aptamers offer significant advantages, owing to their SELEX-based systematic screening method. To fully utilize aptamers for TPD, we designed an aptamer and N-degron ensemble system (AptaGron) that circumvents the need for synthetic conjugations between aptamers and proteolysis-recruiting units. In our AptaGron system, a peptide nucleic acid containing an N-degron peptide and a sequence complementary to the aptamer was designed. Using this system, we successfully degraded three target proteins, tau, nucleolin, and eukaryotic initiation factor 4E (eIF4E), which lack specific small-molecule ligands. Our results highlight the potential of the AptaGron approach as a robust platform for targeted protein degradation.
Collapse
Affiliation(s)
- Mohammad Faysal Al Mazid
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) & Department of Biological Chemistry, KIST School UST, Seoul 02792, South Korea
- Department of Pharmacology, College of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Olha Shkel
- Brain Science Institute, Korea Institute of Science and Technology (KIST) & Department of Biological Chemistry, KIST School UST, Seoul 02792, South Korea
- Department of Pharmacology, College of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Eunteg Ryu
- Department of Pharmacology, College of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Jiwon Kim
- Department of Pharmacology, College of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Kyung Ho Shin
- Department of Pharmacology, College of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Yun Kyung Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST) & Department of Biological Chemistry, KIST School UST, Seoul 02792, South Korea
| | - Hyun Suk Lim
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, South Korea
| | - Jun-Seok Lee
- Department of Pharmacology, College of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, South Korea
| |
Collapse
|
26
|
Naresh GKRS, Guruprasad L. In silico design, modelling and molecular mechanisms of Axl receptor tyrosine kinase inhibitors. J Biomol Struct Dyn 2024:1-14. [PMID: 39681465 DOI: 10.1080/07391102.2024.2439578] [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: 11/14/2023] [Accepted: 04/04/2024] [Indexed: 12/18/2024]
Abstract
A kinase domain from receptor tyrosine kinases (RTKs) regulate intracellular communications to control cellular metabolic activities. Some of the malignant cells have upregulated and overexpressed RTKs which are responsible for angiogenesis in many metastatic cancers. Axl RTK is present in most of the eukaryotic cells and all metastatic cancer cells have overexpressed Axl tyrosine kinase to trigger uncontrolled growth and angiogenesis in the malignant cells. The upregulated kinases can be inhibited in its active and inactive states in the presence of small organic molecule inhibitors. Kinase inhibitors have been discovered to arrest the signal transduction pathways in the malignant cells as a therapy and cure for cancer. In this work, small molecule databases were screened using the pharmacophore features of a macrocyclic inhibitor (7YS) taken as reference from the crystal structure of Axl kinase domain. Pharmacophore based virtual screening of small molecule libraries (CHEMBL32, ChemDiv, Chemspace, Mcule, MolProt, PubChem and Zinc), followed by molecular docking, molecular dynamics simulations, binding energies from MM-PBSA calculations and trajectory analysis as principal component analysis were studied. The molecular basis for the binding of macrocyclic inhibitor, ATP and seven screened hit molecules bound at Axl kinase domain in two different modes at catalytic and regulatory sites was analyzed.
Collapse
|
27
|
Chen J, Fasihianifard P, Lian R, Gibson-Elias LJ, Moreno JL, Chang CEA, Zhong W, Hooley RJ. Supramolecular Host:Guest Arrays Site-Selectively Recognize Peptide Phosphorylation and Kinase Activity. J Am Chem Soc 2024. [PMID: 39680592 DOI: 10.1021/jacs.4c13757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
A synergistic combination of cationic styrylpyridinium dyes and water-soluble deep cavitand hosts can recognize phosphorylated peptides with both site- and state-selectivity. Two mechanisms of interaction are dominant: either the cationic dye interacts with Trp residues in the peptide or the host:dye pair forms a heteroternary complex with the peptide, driven by both strong dye-peptide and cavitand-peptide binding (Kd values up to 4 μM). The presence of multiple recognition mechanisms results in varying fluorescence responses dependent on the phosphorylation state and position, eliminating the need for covalent modification of the peptide target. Differential sensing aided by machine learning algorithms permits full discrimination between differently positioned serine phosphorylations with a minimal 3-component array. The array is fully functional in the presence of protein kinase A (PKA) and its required cofactors and capable of site-selective monitoring of serine phosphorylation at the privileged PKA motif, in the presence of serine residues that do not undergo reaction, illustrating the potential of the system in kinase-based drug screening.
Collapse
Affiliation(s)
- Junyi Chen
- Department of Chemistry, University of California─Riverside, Riverside, California 92521, United States
| | - Parisa Fasihianifard
- Department of Chemistry, University of California─Riverside, Riverside, California 92521, United States
| | - Ria Lian
- Department of Chemistry, University of California─Riverside, Riverside, California 92521, United States
| | - Lucas J Gibson-Elias
- Department of Chemistry, University of California─Riverside, Riverside, California 92521, United States
| | - Jose L Moreno
- Department of Chemistry, University of California─Riverside, Riverside, California 92521, United States
| | - Chia-En A Chang
- Department of Chemistry, University of California─Riverside, Riverside, California 92521, United States
| | - Wenwan Zhong
- Key Laboratory of Precision and Intelligent Chemistry; Department of Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Richard J Hooley
- Department of Chemistry, University of California─Riverside, Riverside, California 92521, United States
| |
Collapse
|
28
|
Wittlinger F, Chitnis SP, Pham CD, Damghani T, Patel KB, Möllers M, Schaeffner IK, Abidakun OA, Deng MQ, Ogboo BC, Rasch A, Beyett TS, Buckley B, Feru F, Shaurova T, Knappe C, Eck MJ, Hershberger PA, Scott DA, Brandt AL, Laufer SA, Heppner DE. Tilting the Scales toward EGFR Mutant Selectivity: Expanding the Scope of Bivalent "Type V" Kinase Inhibitors. J Med Chem 2024; 67:21438-21469. [PMID: 39626019 DOI: 10.1021/acs.jmedchem.4c02311] [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: 12/13/2024]
Abstract
Binding multiple sites within proteins with bivalent compounds is a strategy for developing uniquely active agents. A new class of dual-site inhibitors has emerged targeting the epidermal growth factor receptor (EGFR) anchored to both the orthosteric (ATP) and allosteric sites. Despite proof-of-concept successes, enabling selectivity against oncogenic activating mutations has not been achieved and classifying these inhibitors among kinase inhibitors remains underexplored. This study investigates the structure-activity relationships, binding modes, and biological activity of ATP-allosteric bivalent inhibitors (AABIs). We find that AABIs selectively inhibit drug-resistant EGFR mutants (L858R/T790M and L858R/T790M/C797S) by anchoring a methyl isoindolinone moiety along the αC-helix channel of the allosteric site. In contrast, related Type I1/2 inhibitors target wild-type EGFR but are less effective against resistant mutants. This shift in selectivity demonstrates that mutant-selective AABIs classify as "Type V" bivalent inhibitors.
Collapse
Affiliation(s)
- Florian Wittlinger
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Surbhi P Chitnis
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Calvin D Pham
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Tahereh Damghani
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Kishan B Patel
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Mareike Möllers
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Ilse K Schaeffner
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Omobolanle A Abidakun
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Matthew Q Deng
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Blessing C Ogboo
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Alexander Rasch
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Tyler S Beyett
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Brian Buckley
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, United States
| | - Frederic Feru
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Tatiana Shaurova
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, United States
| | - Cornelius Knappe
- Institute of Pharmaceutical Sciences, Pharmaceutical (Bio-)Analysis, University of Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Michael J Eck
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Pamela A Hershberger
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, United States
| | - David A Scott
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Asher L Brandt
- Department of Chemistry, University of Saint Joseph, West Hartford, Connecticut 06117 United States
| | - Stefan A Laufer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies" Eberhard Karls Universität Tübingen, 72076 Tübingen, Germany
- Tübingen Center for Academic Drug Discovery & Development (TüCAD2), 72076 Tübingen, Germany
| | - David E Heppner
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, United States
- Department of Structural Biology, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| |
Collapse
|
29
|
Pallasaho S, Gondane A, Kutz J, Liang J, Yalala S, Duveau DY, Pospiech H, Thomas CJ, Loda M, Itkonen HM. Compromised CDK12 activity causes dependency on the high activity of O-GlcNAc transferase. Glycobiology 2024; 34:cwae081. [PMID: 39361894 PMCID: PMC11632362 DOI: 10.1093/glycob/cwae081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/19/2024] [Accepted: 10/01/2024] [Indexed: 10/05/2024] Open
Abstract
O-GlcNAc transferase (OGT) coordinates with regulators of transcription, including cyclin-dependent kinase 12 (CDK12), the major transcription elongation kinase. Here, we use inhibitor- and knockdown-based strategies to show that co-targeting of OGT and CDK12 is toxic to prostate cancer cells. OGT catalyzes all nucleocytoplasmic O-GlcNAcylation and due to its essentiality in higher eukaryotes, it is not an ideal drug target. Our glycoproteomics-data revealed that short-term CDK12 inhibition induces hyper-O-GlcNAcylation of the spliceosome-machinery in different models of prostate cancer. By integrating our glycoproteomics-, gene essentiality- and clinical-data from CDK12 mutant prostate cancer patients, we identify the non-essential serine-arginine protein kinase 1 (SRPK1) as a synthetic lethal partner with CDK12-inactivation. Both normal and cancer cells become highly sensitive against inhibitors of OGT and SRPK1 if they have lowered activity of CDK12. Inactivating mutations in CDK12 are enriched in aggressive prostate cancer, and we propose that these patients would benefit from therapy targeting the spliceosome.
Collapse
Affiliation(s)
- Satu Pallasaho
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Aishwarya Gondane
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Julia Kutz
- Project group Biochemistry, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena D-07745, Germany
| | - Jing Liang
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Shivani Yalala
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Damien Y Duveau
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD 20850, United States
| | - Helmut Pospiech
- Project group Biochemistry, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena D-07745, Germany
- University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Life Science Center, Düsseldorf D-40225, Germany
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD 20850, United States
- Lymphoid Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, United States
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York 10021, United States
- The Broad Institute of Harvard and MIT, Cambridge, MA 02142, United States
- The New York Genome Center, New York, New York 10013, United States
| | - Harri M Itkonen
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York 10021, United States
| |
Collapse
|
30
|
Schaller DA, Christ CD, Chodera JD, Volkamer A. Benchmarking Cross-Docking Strategies in Kinase Drug Discovery. J Chem Inf Model 2024; 64:8848-8858. [PMID: 39558632 DOI: 10.1021/acs.jcim.4c00905] [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: 11/20/2024]
Abstract
In recent years, machine learning has transformed many aspects of the drug discovery process, including small molecule design, for which the prediction of bioactivity is an integral part. Leveraging structural information about the interactions between a small molecule and its protein target has great potential for downstream machine learning scoring approaches but is fundamentally limited by the accuracy with which protein-ligand complex structures can be predicted in a reliable and automated fashion. With the goal of finding practical approaches to generating useful kinase-inhibitor complex geometries for downstream machine learning scoring approaches, we present a kinase-centric docking benchmark assessing the performance of different classes of docking and pose selection strategies to assess how well experimentally observed binding modes are recapitulated in a realistic cross-docking scenario. The assembled benchmark data set focuses on the well-studied protein kinase family and comprises a subset of 589 protein structures cocrystallized with 423 ATP-competitive ligands. We find that the docking methods biased by the cocrystallized ligand, utilizing shape overlap with or without maximum common substructure matching, are more successful in recovering binding poses than standard physics-based docking alone. Also, docking into multiple structures significantly increases the chance of generating a low root-mean-square deviation (RMSD) docking pose. Docking utilizing an approach that combines all three methods (Posit) into structures with the most similar cocrystallized ligands according to the maximum common substructure (MCS) proved to be the most efficient way to reproduce binding poses, achieving a success rate of 70.4% across all included systems. The studied docking and pose selection strategies, which utilize the OpenEye Toolkits, were implemented into pipelines of the KinoML framework, allowing automated and reliable protein-ligand complex generation for future downstream machine learning tasks. Although focused on protein kinases, we believe that the general findings can also be transferred to other protein families.
Collapse
Affiliation(s)
- David A Schaller
- In Silico Toxicology and Structural Bioinformatics, Institute of Physiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Clara D Christ
- Structural Biology & Computational Design, Drug Discovery Sciences, Pharmaceuticals, Bayer AG, 13342 Berlin, Germany
| | - John D Chodera
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Andrea Volkamer
- In Silico Toxicology and Structural Bioinformatics, Institute of Physiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Data Driven Drug Design, Faculty of Mathematics and Computer Sciences, Saarland University, 66123 Saarbrücken, Germany
| |
Collapse
|
31
|
Manoharan S, Perumal E. Chemotherapeutic potential of radotinib against blood and solid tumors: A beacon of hope in drug repurposing. Bioorg Chem 2024; 154:108017. [PMID: 39647393 DOI: 10.1016/j.bioorg.2024.108017] [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: 08/15/2024] [Revised: 11/08/2024] [Accepted: 11/28/2024] [Indexed: 12/10/2024]
Abstract
Tyrosine kinase inhibitors (TKIs) represent a pivotal class of targeted therapies in oncology, with multiple generations developed to address diverse molecular targets. Imatinib is the first TKI developed to target the BCR-ABL1 chimeric protein, which is the key driver oncogene implicated in Philadelphia chromosome-positive chronic myeloid leukemia (CML). Several second-generation tyrosine kinase inhibitors (2GTKIs), such as nilotinib, dasatinib, bosutinib, and radotinib (RTB), followed the groundbreaking introduction of imatinib. RTB occupies the unique position of being the least explored member of this class. While nilotinib, dasatinib, and bosutinib have garnered significant attention and extensive research focus, RTB remains relatively uncharted in comparison to its counterparts. Fundamental drug characteristics, such as the pharmacokinetic and pharmacodynamic properties of RTB, remain unavailable in existing sources. Compared to other 2GTKIs, RTB has been less utilized in combinatorial drug studies, and no investigations have been reported on its effects on solid tumors to date. However, the effects of RTB have been studied in acute myeloid leukemia (AML), multiple myeloma (MM), Parkinson's disease, and idiopathic pulmonary fibrosis (IPF). Although RTB has been investigated in some conditions, these studies are still in their preliminary stages and are comparatively lesser than studies on other 2GTKIs. This review is the first attempt that extensively presents a compilation of data on RTB and describes its therapeutic potential against blood and solid tumors. Further investigations on RTB could expand its chemotherapeutic usage in various solid tumors and enhance the possibility of drug repurposing in cancer therapy.
Collapse
Affiliation(s)
- Suryaa Manoharan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641046, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641046, India.
| |
Collapse
|
32
|
Baro M, Lee H, Kelley V, Lou R, Phoomak C, Politi K, Zeiss CJ, Van Zandt M, Contessa JN. OST Catalytic Subunit Redundancy Enables Therapeutic Targeting of N-Glycosylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626593. [PMID: 39677793 PMCID: PMC11643024 DOI: 10.1101/2024.12.03.626593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Protein asparagine (N)-glycosylation, which promotes folding and trafficking of cell surface receptors such as the EGFR, has not been considered a viable target in oncology due to the essential and non-redundant enzymatic activities required for glycan synthesis and transfer. In mammals an exception to this rule is the presence of the oligosaccharyltransferase (OST) catalytic subunit paralogs, STT3A and STT3B. Here we delineate the chemical biology of OST inhibitors and develop an approach for limited inhibition of N-glycosylation optimized for downstream effects on EGFR. Small molecules with enhanced pharmacokinetic properties and preferences for STT3A or STT3B were synthesized, characterized in vitro, and advanced to in vivo testing. The lead from this series, NGI-189, causes tumor regression or growth delay of patient derived and TKI resistant EGFR-mutant lung cancer xenografts without toxicity. Together these results suggest that bioavailable OST inhibitors can be developed as therapeutic agents for oncology.
Collapse
Affiliation(s)
- Marta Baro
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06510 USA
| | - Hojin Lee
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06510 USA
| | - Vanessa Kelley
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06510 USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06510 USA
| | - Rongliang Lou
- New England Discovery Partners, Branford, CT, 06405 USA
| | - Chatchai Phoomak
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06510 USA
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok 10330 Thailand
| | - Katerina Politi
- Department of Internal Medicine (Section of Medical Oncology), Yale University School of Medicine, New Haven, CT, 06510 USA
| | - Caroline J. Zeiss
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Joseph N. Contessa
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06510 USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06510 USA
- Lead contact
| |
Collapse
|
33
|
Estevam GO, Linossi EM, Rao J, Macdonald CB, Ravikumar A, Chrispens KM, Capra JA, Coyote-Maestas W, Pimentel H, Collisson EA, Jura N, Fraser JS. Mapping kinase domain resistance mechanisms for the MET receptor tyrosine kinase via deep mutational scanning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603579. [PMID: 39071407 PMCID: PMC11275805 DOI: 10.1101/2024.07.16.603579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Mutations in the kinase and juxtamembrane domains of the MET Receptor Tyrosine Kinase are responsible for oncogenesis in various cancers and can drive resistance to MET-directed treatments. Determining the most effective inhibitor for each mutational profile is a major challenge for MET-driven cancer treatment in precision medicine. Here, we used a deep mutational scan (DMS) of ~5,764 MET kinase domain variants to profile the growth of each mutation against a panel of 11 inhibitors that are reported to target the MET kinase domain. We validate previously identified resistance mutations, pinpoint common resistance sites across type I, type II, and type I ½ inhibitors, unveil unique resistance and sensitizing mutations for each inhibitor, and verify non-cross-resistant sensitivities for type I and type II inhibitor pairs. We augment a protein language model with biophysical and chemical features to improve the predictive performance for inhibitor-treated datasets. Together, our study demonstrates a pooled experimental pipeline for identifying resistance mutations, provides a reference dictionary for mutations that are sensitized to specific therapies, and offers insights for future drug development.
Collapse
Affiliation(s)
- Gabriella O. Estevam
- Department of Bioengineering and Therapeutic Sciences, UCSF, San Francisco, CA, United States
- Tetrad Graduate Program, UCSF, San Francisco, CA, United States
| | - Edmond M. Linossi
- Cardiovascular Research Institute, UCSF, San Francisco, CA, United States
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA, United States
| | - Jingyou Rao
- Department of Computer Science, UCLA, Los Angeles, CA, United States
| | - Christian B. Macdonald
- Department of Bioengineering and Therapeutic Sciences, UCSF, San Francisco, CA, United States
| | - Ashraya Ravikumar
- Department of Bioengineering and Therapeutic Sciences, UCSF, San Francisco, CA, United States
| | - Karson M. Chrispens
- Department of Bioengineering and Therapeutic Sciences, UCSF, San Francisco, CA, United States
- Biophysics Graduate Program, UCSF, San Francisco, CA, United States
| | - John A. Capra
- Bakar Computational Health Sciences Institute and Department of Epidemiology and Biostatistics, UCSF, San Francisco, CA, United States
| | - Willow Coyote-Maestas
- Department of Bioengineering and Therapeutic Sciences, UCSF, San Francisco, CA, United States
- Quantitative Biosciences Institute, UCSF, San Francisco, CA, United States
| | - Harold Pimentel
- Department of Computer Science, UCLA, Los Angeles, CA, United States
- Department of Computational Medicine and Human Genetics, UCLA, Los Angeles, CA, United States
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| | - Eric A. Collisson
- Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States
- Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Natalia Jura
- Cardiovascular Research Institute, UCSF, San Francisco, CA, United States
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA, United States
- Quantitative Biosciences Institute, UCSF, San Francisco, CA, United States
| | - James S. Fraser
- Department of Bioengineering and Therapeutic Sciences, UCSF, San Francisco, CA, United States
- Quantitative Biosciences Institute, UCSF, San Francisco, CA, United States
| |
Collapse
|
34
|
Ritika, Liao ZY, Chen PY, Rao NV, Mathew J, Sharma R, Grewal AS, Singh G, Mehan S, Liou JP, Pan CH, Nepali K. Rationally designed febuxostat-based hydroxamic acid and its pH-Responsive nanoformulation elicits anti-tumor activity. Eur J Med Chem 2024; 279:116866. [PMID: 39293244 DOI: 10.1016/j.ejmech.2024.116866] [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: 05/21/2024] [Revised: 08/29/2024] [Accepted: 09/08/2024] [Indexed: 09/20/2024]
Abstract
Attempts to furnish antitumor structural templates that can prevent the occurrence of drug-induced hyperuricemia spurred us to generate xanthine oxidase inhibitor-based hydroxamic acids and anilides. Specifically, the design strategy involved the insertion of febuxostat (xanthine oxidase inhibitor) as a surface recognition part of the HDAC inhibitor pharmacophore model. Investigation outcomes revealed that hydroxamic acid 4 elicited remarkable antileukemic effects mediated via HDAC isoform inhibition. Delightfully, the adduct retained xanthine oxidase inhibitory activity, though xanthine oxidase inhibition was not the underlying mechanism of its cell growth inhibitory effects. Also, compound 4 demonstrated significant in-vivo anti-hyperuricemic (PO-induced hyperuricemia model) and antitumor activity in an HL-60 xenograft mice model. Compound 4 was conjugated with poly (ethylene glycol) poly(aspartic acid) block copolymer to furnish pH-responsive nanoparticles (NPs) in pursuit of circumventing its cytotoxicity towards the normal cell lines. SEM analysis revealed that NPs had uniform size distributions, while TEM analysis ascertained the spherical shape of NPs, indicating their ability to undergo self-assembly. HDAC inhibitor 4 was liberated from the matrix due to the polymeric nanoformulation's pH-responsiveness, and the NPs demonstrated selective cancer cell targeting ability.
Collapse
Affiliation(s)
- Ritika
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taiwan
| | - Zi-Yi Liao
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110031, Taiwan
| | - Pin-Yu Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110031, Taiwan
| | - N Vijayakamasewara Rao
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan
| | - Jacob Mathew
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 106335, Taiwan
| | - Ram Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110031, Taiwan
| | - Ajmer Singh Grewal
- Department of Pharmaceutical Sciences, Guru Gobind Singh College of Pharmacy, Near Guru Nanak Khalsa College, Yamuna Nagar, 135001, Haryana, India
| | - Gurpreet Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India; Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Jing Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110031, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan
| | - Chun Hsu Pan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan.
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110031, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan.
| |
Collapse
|
35
|
Huang C, Huang X, Qiu X, Kong X, Wu C, Jiang X, Yao M, Wang M, Su L, Lv C, Wong P. Pericytes Modulate Third-Generation Tyrosine Kinase Inhibitor Sensitivity in EGFR-Mutated Lung Cancer Cells Through IL32-β5-Integrin Paracrine Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405130. [PMID: 39435643 PMCID: PMC11633494 DOI: 10.1002/advs.202405130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/17/2024] [Indexed: 10/23/2024]
Abstract
EGFR-mutated lung cancer patients sometimes display restricted responses to third-generation tyrosine kinase inhibitors (TKIs), potentially attributable to undervalued input from stromal cells, notably pericytes (PCs). The study shows that PCs isolated from EGFR-mutated patients have a unique secretome profile, notably secreting IL32 and affecting signaling pathways and biological processes linked to TKI sensitivity. Clinical evidence, supported by single-cell RNA sequencing and multiplex immunostaining of tumor tissues, confirms the presence of IL32-expressing pericytes closely interacting with β5-integrin-expressing cancer cells in EGFR-mutated patients, impacting therapeutic response and prognosis. Co-culture and conditioned medium experiments demonstrate that PCs reduce TKI effectiveness in EGFR-mutated cancer cells, a reversible phenomenon through silencing IL32 expression in PCs or depleting the IL32 receptor β5-integrin on cancer cells, thereby restoring cancer cell sensitivity. Mechanistically, it is shown that YY1 signaling upregulates IL32 secretion in PCs, subsequently activating the β5-integrin-Src-Akt pathway in EGFR-mutated cancer cells, contributing to their TKI sensitivity. In animal studies, co-injection of cancer cells with PCs compromises TKI effectiveness, independently of blood vessel functions, while inhibition of β5-integrin restores tumor cell sensitivity. Overall, the findings highlight direct crosstalk between cancer cells and pericytes, impacting TKI sensitivity via IL32-β5-integrin paracrine signaling, proposing an enhanced therapeutic approach for EGFR-mutated patients.
Collapse
Affiliation(s)
- Cheng Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Guangzhou Key Laboratory of Precise Diagnosis and Treatment of Biliary Tract CancerDepartment of Biliary‐Pancreatic SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Xi Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Xiaoyi Qiu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Xiangzhan Kong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Chunmiao Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Xue Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Mingkang Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Department of Respiratory MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Minghui Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Department of Thoracic SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Liangping Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Guangdong Provincial Key Laboratory of Urological DiseasesGuangzhou Medical UniversityGuangzhou510120China
| | - Cui Lv
- Clinical Biobank CenterZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Ping‐Pui Wong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Guangzhou Key Laboratory of Precise Diagnosis and Treatment of Biliary Tract CancerDepartment of Biliary‐Pancreatic SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| |
Collapse
|
36
|
Hill AD, Okonechnikov K, Herr MK, Thomas C, Thongjuea S, Hasselblatt M, Patrizi A. Single-nucleus RNA-seq dissection of choroid plexus tumor cell heterogeneity. EMBO J 2024; 43:6766-6791. [PMID: 39482394 PMCID: PMC11649822 DOI: 10.1038/s44318-024-00283-2] [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: 10/07/2023] [Revised: 10/08/2024] [Accepted: 10/15/2024] [Indexed: 11/03/2024] Open
Abstract
The genomic, genetic and cellular events regulating the onset, growth and survival of rare, choroid plexus neoplasms remain poorly understood. Here, we examine the heterogeneity of human choroid plexus tumors by single-nucleus transcriptome analysis of 23,906 cells from four disease-free choroid plexus and eleven choroid plexus tumors. The resulting expression atlas profiles cellular and transcriptional diversity, copy number alterations, and cell-cell interaction networks in normal and cancerous choroid plexus. In choroid plexus tumor epithelial cells, we observe transcriptional changes that correlate with genome-wide methylation profiles. We further characterize tumor type-specific stromal microenvironments that include altered macrophage and mesenchymal cell states, as well as changes in extracellular matrix components. This first single-cell dataset resource from such scarce samples should be valuable for divising therapies against these little-studied neoplasms.
Collapse
Affiliation(s)
- Anthony D Hill
- Schaller Research Group, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.
| | - Konstantin Okonechnikov
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
| | - Marla K Herr
- Schaller Research Group, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177, Stockholm, Sweden
| | - Christian Thomas
- Institute of Neuropathology, University Hospital Münster, 48149, Münster, Germany
| | - Supat Thongjuea
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
| | - Martin Hasselblatt
- Institute of Neuropathology, University Hospital Münster, 48149, Münster, Germany
| | - Annarita Patrizi
- Schaller Research Group, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.
| |
Collapse
|
37
|
Howard PG, Zou P, Zhang Y, Huang F, Tesic V, Wu CYC, Lee RHC. Serum/glucocorticoid regulated kinase 1 (SGK1) in neurological disorders: pain or gain. Exp Neurol 2024; 382:114973. [PMID: 39326820 PMCID: PMC11536509 DOI: 10.1016/j.expneurol.2024.114973] [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: 08/26/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024]
Abstract
Serum/Glucocorticoid Regulated Kinase 1 (SGK1), a serine/threonine kinase, is ubiquitous across a wide range of tissues, orchestrating numerous signaling pathways and associated with various human diseases. SGK1 has been extensively explored in diverse types of immune and inflammatory diseases, cardiovascular disorders, as well as cancer metastasis. These studies link SGK1 to cellular proliferation, survival, metabolism, membrane transport, and drug resistance. Recently, increasing research has focused on SGK1's role in neurological disorders, including a variety of neurodegenerative diseases (e.g., Alzheimer's disease, Huntington's disease and Parkinson's disease), brain injuries (e.g., cerebral ischemia and traumatic brain injury), psychiatric conditions (e.g., depression and drug addiction). SGK1 is emerging as an increasingly compelling therapeutic target across the spectrum of neurological disorders, supported by the availability of several effective agents. However, the conclusions of many studies observing the prevalence and function of SGK1 in neurological disorders are contradictory, necessitating a review of the SGK1 research within neurological disorders. Herein, we review recent literature on SGK1's primary functions within the nervous system and its impacts within different neurological disorders. We summarize significant findings, identify research gaps, and outline possible future research directions based on the current understanding of SGK1 to help further progress the understanding and treatment of neurological disorders.
Collapse
Affiliation(s)
- Peyton Grace Howard
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Peibin Zou
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Yulan Zhang
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Fang Huang
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Vesna Tesic
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Celeste Yin-Chieh Wu
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA.
| | - Reggie Hui-Chao Lee
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA; Department of Department of Cell Biology & Anatomy, Louisiana State University Health, Shreveport, LA, USA.
| |
Collapse
|
38
|
Olszewski M, Stasevych M, Zvarych V, Maciejewska N. 9,10-Dioxoanthracenyldithiocarbamates effectively inhibit the proliferation of non-small cell lung cancer by targeting multiple protein tyrosine kinases. J Enzyme Inhib Med Chem 2024; 39:2284113. [PMID: 38078360 PMCID: PMC11721965 DOI: 10.1080/14756366.2023.2284113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/26/2023] [Accepted: 11/12/2023] [Indexed: 12/18/2023] Open
Abstract
Anthraquinones have attracted considerable interest in the realm of cancer treatment owing to their potent anticancer properties. This study evaluates the potential of a series of new anthraquinone derivatives as anticancer agents for non-small-cell lung cancer (NSCLC). The compounds were subjected to a range of tests to assess their cytotoxic and apoptotic properties, ability to inhibit colony formation, pro-DNA damage functions, and capacity to inhibit the activity of tyrosine kinase proteins (PTKs). Based on the research findings, it has been discovered that most active derivatives (i84, i87, and i90) possess a substantial capability to impede the viability of NSCLC while having mostly a negligible effect on the human kidney cell line. Moreover, the anthraquinones displayed pro-apoptotic and genotoxic attributes while blocking the phosphorylation of multiple PTKs. Collectively, our findings indicate that these derivatives may demonstrate promising potential as effective anticancer agents for lung cancer treatment.
Collapse
Affiliation(s)
- Mateusz Olszewski
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Maryna Stasevych
- Department of Technology of Biologically Active Substances, Pharmacy, and Biotechnology, Lviv Polytechnic National University 13, Lviv, Ukraine
| | - Viktor Zvarych
- Department of Technology of Biologically Active Substances, Pharmacy, and Biotechnology, Lviv Polytechnic National University 13, Lviv, Ukraine
| | - Natalia Maciejewska
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| |
Collapse
|
39
|
Mukherjee S, Joshi V, Reddy KP, Singh N, Das P, Datta P. Biopharmaceutical and pharmacokinetic attributes to drive nanoformulations of small molecule tyrosine kinase inhibitors. Asian J Pharm Sci 2024; 19:100980. [PMID: 39640056 PMCID: PMC11617995 DOI: 10.1016/j.ajps.2024.100980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 06/13/2024] [Accepted: 06/29/2024] [Indexed: 12/07/2024] Open
Abstract
Buoyed by the discovery of small-molecule tyrosine kinase inhibitors (smTKIs), significant impact has been made in cancer chemotherapeutics. However, some of these agents still encounter off-target toxicities and suboptimal efficacies due to their inferior biopharmaceutical and/or pharmacokinetic properties. Almost all of these molecules exhibit significant inter- and intra-patient variations in plasma concentration-time profiles. Thus, therapeutic drug monitoring, dose adjustments and precision medicine are being contemplated by clinicians. Complex formulations or nanoformulation-based drug delivery systems offer promising approaches to provide drug encapsulation or spatiotemporal control over the release, overcoming the biopharmaceutical and pharmacokinetic limitations and improving the therapeutic outcomes. In this context, the present review comprehensively tabulates and critically analyzes all the relevant properties (T1/2, solubility, pKa, therapeutic index, IC50, metabolism etc.) of the approved smTKIs. A detailed appraisal is conducted on the advancements made in complex formulations of smTKIs, with a focus on strategies to enhance their pharmacokinetic profile, tumor targeting ability, and therapeutic efficacy. Various nanocarrier platforms, have been discussed, highlighting their unique features and potential applications in cancer therapy. Nanoformulations have been shown to improve area under the curve and peak plasma concentration, and reduce dosing frequency for several smTKIs in animal models. It is inferred that extensive efforts will be made in developing complex formulations of smTKIs in near future. There, the review concludes with key recommendations for the developing of smTKIs to facilitate early clinical translation.
Collapse
Affiliation(s)
| | | | - Kolimi Prashanth Reddy
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| | - Nidhi Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| | - Priyanka Das
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| | - Pallab Datta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| |
Collapse
|
40
|
Cheng SS, Mody AC, Woo CM. Opportunities for Therapeutic Modulation of O-GlcNAc. Chem Rev 2024; 124:12918-13019. [PMID: 39509538 DOI: 10.1021/acs.chemrev.4c00417] [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: 11/15/2024]
Abstract
O-Linked β-N-acetylglucosamine (O-GlcNAc) is an essential, dynamic monosaccharide post-translational modification (PTM) found on serine and threonine residues of thousands of nucleocytoplasmic proteins. The installation and removal of O-GlcNAc is controlled by a single pair of enzymes, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), respectively. Since its discovery four decades ago, O-GlcNAc has been found on diverse classes of proteins, playing important functional roles in many cellular processes. Dysregulation of O-GlcNAc homeostasis has been implicated in the pathogenesis of disease, including neurodegeneration, X-linked intellectual disability (XLID), cancer, diabetes, and immunological disorders. These foundational studies of O-GlcNAc in disease biology have motivated efforts to target O-GlcNAc therapeutically, with multiple clinical candidates under evaluation. In this review, we describe the characterization and biochemistry of OGT and OGA, cellular O-GlcNAc regulation, development of OGT and OGA inhibitors, O-GlcNAc in pathophysiology, clinical progress of O-GlcNAc modulators, and emerging opportunities for targeting O-GlcNAc. This comprehensive resource should motivate further study into O-GlcNAc function and inspire strategies for therapeutic modulation of O-GlcNAc.
Collapse
Affiliation(s)
- Steven S Cheng
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Alison C Mody
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Christina M Woo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
- Affiliate member of the Broad Institute, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
41
|
Woods K, Rants'o TA, Chan AM, Sapre T, Mastin GE, Maguire KM, Ong SE, Golkowski M. diaPASEF-Powered Chemoproteomics Enables Deep Kinome Interaction Profiling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624841. [PMID: 39605566 PMCID: PMC11601655 DOI: 10.1101/2024.11.22.624841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Protein-protein interactions (PPIs) underlie most biological functions. Devastating human conditions like cancers, neurological disorders, and infections, hijack PPI networks to initiate disease, and to drive disease progression. Understanding precisely how diseases remodel PPI networks can, therefore, help clarify disease mechanisms and identify therapeutic targets. Protein kinases control most cellular processes through protein phosphorylation. The 518 human kinases, known as the kinome, are frequently dysregulated in disease and highly druggable with ATP-competitive inhibitors. Kinase activity, localization, and substrate recognition are regulated by dynamic PPI networks composed of scaffolding and adapter proteins, other signaling enzymes like small GTPases and E3 ligases, and phospho-substrates. Accordingly, mapping kinase PPI networks can help determine kinome activation states, and, in turn, cellular activation states; this information can be used for studying kinase-mediated cell signaling, and for prioritizing kinases for drug discovery. Previously, we have developed a high-throughput method for kinome PPI mapping based on mass spectrometry (MS)-based chemoproteomics that we named kinobead competition and correlation analysis (kiCCA). Here, we introduce 2 nd generation (gen) kiCCA which utilizes data-independent acquisition (dia) with parallel accumulation serial fragmentation (PASEF) MS and a re-designed CCA algorithm with improved selection criteria and the ability to predict multiple kinase interaction partners of the same proteins. Using neuroblastoma cell line models of the noradrenergic-mesenchymal transition (NMT), we demonstrate that 2 nd gen kiCCA (1) identified 6.1-times more kinase PPIs in native cell extracts compared to our 1 st gen approach, (2) determined kinase-mediated signaling pathways that underly the neuroblastoma NMT, and (3) accurately predicted pharmacological targets for manipulating NMT states. Our 2 nd gen kiCCA method is broadly useful for cell signaling research and kinase drug discovery.
Collapse
|
42
|
Rosenthal KJ, Gordan JD, Scott JD. Protein kinase A and local signaling in cancer. Biochem J 2024; 481:1659-1677. [PMID: 39540434 DOI: 10.1042/bcj20230352] [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: 09/23/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Protein kinase A (PKA) is a basophilic kinase implicated in the modulation of many cell-signaling and physiological processes. PKA also contributes to cancer-relevant events such as growth factor action, cell cycle control, cell migration and tumor metabolism. Germline and somatic mutations in PKA, gene amplifications, and chromosome rearrangements that encode kinase fusions, are linked to a growing number of malignant neoplasms. Mislocalization of PKA by exclusion from A-Kinase Anchoring Protein (AKAP) signaling islands further underlies cancer progression. This article highlights the influence of AKAP signaling and local kinase action in selected hallmarks of cancer. We also feature the utility of kinase inhibitor drugs as frontline and future anti-cancer therapies.
Collapse
Affiliation(s)
- Kacey J Rosenthal
- Department of Pharmacology, University of Washington School of Medicine, 1959 NE Pacific St., Box 357750, Seattle, WA 98195, U.S.A
| | - John D Gordan
- Department of Medicine (Hematology/Oncology), Quantitative Biosciences Institute, UCSF Helen Diller Family Cancer Center, 1700 4th St., San Francisco, CA 94143, U.S.A
| | - John D Scott
- Department of Pharmacology, University of Washington School of Medicine, 1959 NE Pacific St., Box 357750, Seattle, WA 98195, U.S.A
| |
Collapse
|
43
|
Hillis AL, Tamir T, Perry GE, Asara JM, Johnson JL, Yaron TM, Cantley LC, White FM, Toker A. Parallel phosphoproteomics and metabolomics map the global metabolic tyrosine phosphoproteome. Proc Natl Acad Sci U S A 2024; 121:e2413837121. [PMID: 39536083 PMCID: PMC11588116 DOI: 10.1073/pnas.2413837121] [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: 07/10/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
Tyrosine phosphorylation of metabolic enzymes is an evolutionarily conserved posttranslational modification that facilitates rapid and reversible modulation of enzyme activity, localization, or function. Despite the high abundance of tyrosine phosphorylation events detected on metabolic enzymes in high-throughput mass spectrometry-based studies, functional characterization of tyrosine phosphorylation sites has been limited to a subset of enzymes. Since tyrosine phosphorylation is dysregulated across human diseases, including cancer, understanding the consequences of metabolic enzyme tyrosine phosphorylation events is critical for informing disease biology and therapeutic interventions. To globally identify metabolic enzyme tyrosine phosphorylation events and simultaneously assign functional significance to these sites, we performed parallel phosphoproteomics and polar metabolomics in nontumorigenic mammary epithelial cells (MCF10A) stimulated with epidermal growth factor (EGF) in the absence or presence of the EGF receptor inhibitor erlotinib. We performed an integrated analysis of the phosphoproteomic and metabolomic datasets to identify tyrosine phosphorylation sites on metabolic enzymes with functional consequences. We identified two previously characterized (pyruvate kinase muscle isozyme, phosphoglycerate mutase 1) and two uncharacterized (glutathione S-transferase Pi 1, glutamate dehydrogenase 1) tyrosine phosphorylation sites on metabolic enzymes with purported functions based on metabolomic analyses. We validated these hits using a doxycycline-inducible CRISPR interference system in MCF10A cells, in which target metabolic enzymes were depleted with simultaneous reexpression of wild-type, phosphomutant, or phosphomimetic isoforms. Together, these data provide a framework for identification, prioritization, and characterization of tyrosine phosphorylation sites on metabolic enzymes with functional significance.
Collapse
Affiliation(s)
- Alissandra L. Hillis
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Tigist Tamir
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Grace E. Perry
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - John M. Asara
- Mass Spectrometry Core, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Jared L. Johnson
- Department of Cell Biology, Harvard Medical School, Boston, MA02115
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
- Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | - Tomer M. Yaron
- Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | - Lewis C. Cantley
- Department of Cell Biology, Harvard Medical School, Boston, MA02115
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
- Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | - Forest M. White
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Alex Toker
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| |
Collapse
|
44
|
Das D, Xie L, Qiao D, Jia J, Hong J. Discovery of novel, orally bioavailable phenylacetamide derivatives as multikinase inhibitors and in vivo efficacy study in hepatocellular carcinoma animal models. Bioorg Med Chem Lett 2024; 113:129971. [PMID: 39306048 DOI: 10.1016/j.bmcl.2024.129971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/07/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024]
Abstract
Hepatocellular carcinoma (HCC) is considered as one of the leading causes of death in liver disease patients. Several signal transduction pathways are involved in HCC pathogenesis. Multikinase inhibitors (MKIs) show beneficial effects for HCC and the FDA approved a few MKIs including sorafenib, lenvatinib for HCC treatments. Here, a novel series of phenylacetamide derivatives were designed, synthesized and evaluated as multikinase inhibitors. Several compounds showed nanomolar IC50 values against FLT1, FLT3, FLT4, KDR, PDGFRα, PDGFRβ. The compounds were tested against human hepatocellular carcinoma (HCC), human colon adenocarcinoma and human gastric carcinoma cell lines. With favorable pharmacokinetics profiles, compound 12 and compound 14 were selected for in vivo efficacy studies in Hep3B mice models and demonstrated efficacious than sorafenib.
Collapse
Affiliation(s)
- Debasis Das
- Arromax Pharmatech Co. Ltd. Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou 215123, PR China.
| | - Lingzhi Xie
- Arromax Pharmatech Co. Ltd. Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou 215123, PR China
| | - Dandan Qiao
- Arromax Pharmatech Co. Ltd. Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou 215123, PR China
| | - Jianhe Jia
- Arromax Pharmatech Co. Ltd. Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou 215123, PR China
| | - Jian Hong
- Arromax Pharmatech Co. Ltd. Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou 215123, PR China.
| |
Collapse
|
45
|
Zheng J, Zhang W, Ni D, Zhao S, He Y, Hu J, Li L, Dang Y, Guo Z, Nie S. Design, Synthesis, and Biological Evaluation of 3-Amino-pyrazine-2-carboxamide Derivatives as Novel FGFR Inhibitors. ACS Med Chem Lett 2024; 15:2019-2031. [PMID: 39563808 PMCID: PMC11571013 DOI: 10.1021/acsmedchemlett.4c00431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/21/2024] Open
Abstract
FGFR has been considered a crucial oncogenic driver and promising target for cancer therapy. Herein, we reported the design and synthesis of 3-amino-N-(3,5-dihydroxyphenyl)-6-methylpyrazine-2-carboxamide derivatives as novel FGFR inhibitors. SAR exploration led to the identification of 18i as a pan-FGFR inhibitor with favorable in vitro activity against FGFR1-4. Moreover, 18i blocked the activation of FGFR and downstream signaling pathways at the submicromolar level and exhibited potent antitumor activity in multiple cancer cell lines with FGFR abnormalities. Molecular docking was performed to investigate the possible binding modes of 18i within the binding site of FGFR2. These results suggest that compound 18i is a promising candidate for further drug discovery.
Collapse
Affiliation(s)
- Jia Zheng
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention (Ministry of Education), College of Pharmacy and Department of Urology of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Wei Zhang
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention (Ministry of Education), College of Pharmacy and Department of Breast and Thyroid Surgery of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Dan Ni
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention (Ministry of Education), College of Pharmacy and Department of Urology of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Shuang Zhao
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention (Ministry of Education), College of Pharmacy and Department of Urology of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yi He
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention (Ministry of Education), College of Pharmacy and Department of Urology of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Junchi Hu
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention (Ministry of Education), College of Pharmacy and Department of Urology of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Linfeng Li
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention (Ministry of Education), College of Pharmacy and Department of Breast and Thyroid Surgery of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yongjun Dang
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention (Ministry of Education), College of Pharmacy and Department of Urology of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Zufeng Guo
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention (Ministry of Education), College of Pharmacy and Department of Breast and Thyroid Surgery of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Shenyou Nie
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention (Ministry of Education), College of Pharmacy and Department of Urology of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
46
|
Echepare M, Picabea B, Arricibita A, Teijeira Á, Pasquier A, Zandueta C, Otegui N, Santamaría E, Fernández-Irigoyen J, Romero O, Sanchez-Cespedes M, Lecanda F, Hernández J, Felip E, Cruz-Bermúdez A, Provencio M, Gentili M, Facchinetti F, Roz L, Montuenga LM, Valencia K. DSTYK Inhibition Sensitizes NSCLC to Taxane-Based Chemotherapy. J Thorac Oncol 2024:S1556-0864(24)02438-9. [PMID: 39536877 DOI: 10.1016/j.jtho.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Chemotherapy continues to be the standard treatment for patients noneligible for targeted or immune-based therapies; nevertheless, treatment resistance remains a major clinical challenge. We previously found that expression levels of DSTYK, a poorly explored dual serine/threonine and tyrosine kinase frequently amplified in cancer, identified patients with lung cancer exhibiting poor response to immune checkpoint inhibitors, and found that its inhibition sensitizes to immunotherapy. Seeking to explore the potential of DSTYK targeting in additional indications, we investigated the functional relevance and actionability of DSTYK in lung cancer chemoresistance. METHODS In silico analysis to study the differential sensitivity to paclitaxel, carboplatin, pemetrexed, and cisplatin drugs was performed using pan-cancer human cancer cell line DSTYK CN data downloaded from Depmap portal (https://depmap.org/portal/depmap/). Two cohorts of patients with lung cancer were used. An adjuvant cohort composed of patients with advanced IV-stage lung adenocarcinoma treated with carboplatin plus paclitaxel or carboplatin plus pemetrexed in the first line from VHIO; A neoadjuvant cohort including resectable stage IIIA or IIIB NSCLC tumor samples of patients from NADIM I and NADIM II clinical trials perioperative treated with nivolumab plus chemotherapy (carboplatin + paclitaxel). DSTYK CN was assessed by fluorescence in situ hybridization and quantitative reverse transcription-polymerase chain reaction. Proteomics and bioinformatic analyses were performed to study differentially expressed protein signatures. Functional in vitro experiments (adhesion, migration, and invasion) in both murine and human systems, and in vivo lung orthotopic, intracardiac, and intratibial xenograft and syngeneic models complete the study. RESULTS We show that DSTYK depletion specifically sensitizes lung cancer cells to taxane-based chemotherapy, particularly in combination with carboplatin. Mechanistically, DSTYK ablation remodels the cytoskeleton and impairs distant invasion and metastatic outgrowth in vivo. DSTYK downregulation sensitizes both primary and metastatic lung tumors to chemoimmunotherapy treatment leading to tumor regression in mouse models. Consistently, clinical data of patients with early and advanced lung cancer-in the neoadjuvant and adjuvant settings-show a strong correlation between DSTYK amplification and taxane resistance, underscoring the clinical significance of our findings to inform treatment decision-making. CONCLUSIONS Collectively, our data indicates that DSTYK amplification may be a predictor of resistance to taxane-based treatments and represents an actionable target for these patients.
Collapse
Affiliation(s)
- Mirari Echepare
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain; Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Beñat Picabea
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Andrea Arricibita
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain
| | - Álvaro Teijeira
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain; Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Navarra Health Research Institute (IDISNA), Pamplona, Spain; Program of Immunology and Immunotherapy, CIMA-University of Navarra, Pamplona, Spain
| | - Andrea Pasquier
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain
| | - Carolina Zandueta
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain
| | - Nerea Otegui
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Enrique Santamaría
- Navarra Health Research Institute (IDISNA), Pamplona, Spain; Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Navarra Health Research Institute (IDISNA), Pamplona, Spain; Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain
| | - Octavio Romero
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | | | - Fernando Lecanda
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain
| | - Javier Hernández
- Department of Pathology, Hospital Universitari Vall d'Hebron & Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Enriqueta Felip
- Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VIHO), Universitat Autonoma de Barcelona, Spain
| | - Alberto Cruz-Bermúdez
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Mariano Provencio
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Marco Gentili
- Tumor Genomics Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federica Facchinetti
- Tumor Genomics Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Luca Roz
- Tumor Genomics Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Luis M Montuenga
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain; Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Karmele Valencia
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain; Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Navarra Health Research Institute (IDISNA), Pamplona, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain.
| |
Collapse
|
47
|
Fiedorczuk K, Iordanov I, Mihályi C, Szollosi A, Csanády L, Chen J. The structures of protein kinase A in complex with CFTR: Mechanisms of phosphorylation and noncatalytic activation. Proc Natl Acad Sci U S A 2024; 121:e2409049121. [PMID: 39495916 PMCID: PMC11573500 DOI: 10.1073/pnas.2409049121] [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: 05/06/2024] [Accepted: 10/03/2024] [Indexed: 11/06/2024] Open
Abstract
Protein kinase A (PKA) is a key regulator of cellular functions by selectively phosphorylating numerous substrates, including ion channels, enzymes, and transcription factors. It has long served as a model system for understanding the eukaryotic kinases. Using cryoelectron microscopy, we present complex structures of the PKA catalytic subunit (PKA-C) bound to a full-length protein substrate, the cystic fibrosis transmembrane conductance regulator (CFTR)-an ion channel vital to human health. CFTR gating requires phosphorylation of its regulatory (R) domain. Unphosphorylated CFTR engages PKA-C at two locations, establishing two "catalytic stations" near to, but not directly involving, the R domain. This configuration, coupled with the conformational flexibility of the R domain, permits transient interactions of the eleven spatially separated phosphorylation sites. Furthermore, we determined two structures of the open-pore CFTR stabilized by PKA-C, providing a molecular basis to understand how PKA-C stimulates CFTR currents even in the absence of phosphorylation.
Collapse
Affiliation(s)
- Karol Fiedorczuk
- Laboratory of Membrane Biology and Biophysics, The Rockefeller University, New York, NY 10065
| | - Iordan Iordanov
- Department of Biochemistry, Semmelweis University, Budapest H-1094, Hungary
- Ion Channel Research Group, Hungarian Research Network - Semmelweis University, Budapest H-1094, Hungary
| | - Csaba Mihályi
- Department of Biochemistry, Semmelweis University, Budapest H-1094, Hungary
- Ion Channel Research Group, Hungarian Research Network - Semmelweis University, Budapest H-1094, Hungary
| | - Andras Szollosi
- Department of Biochemistry, Semmelweis University, Budapest H-1094, Hungary
- Ion Channel Research Group, Hungarian Research Network - Semmelweis University, Budapest H-1094, Hungary
| | - László Csanády
- Department of Biochemistry, Semmelweis University, Budapest H-1094, Hungary
- Ion Channel Research Group, Hungarian Research Network - Semmelweis University, Budapest H-1094, Hungary
| | - Jue Chen
- Laboratory of Membrane Biology and Biophysics, The Rockefeller University, New York, NY 10065
- HHMI, Chevy Chase, MD 20815
| |
Collapse
|
48
|
Noguchi M, Miyauchi A, Masaki Y, Sakaki M, Lei XF, Kobayashi-Tanabe M, Miyazaki A, Aoki T, Yoshida H, Seio K, Kim-Kaneyama JR. Hic-5 antisense oligonucleotide inhibits advanced hepatic fibrosis and steatosis in vivo. JHEP Rep 2024; 6:101195. [PMID: 39444410 PMCID: PMC11497448 DOI: 10.1016/j.jhepr.2024.101195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 07/27/2024] [Accepted: 08/05/2024] [Indexed: 10/25/2024] Open
Abstract
Background & Aims Chronic liver diseases, including metabolic dysfunction-associated steatohepatitis (MASH), pose a significant global health burden. Progressive liver fibrosis can lead to severe outcomes; however, there is a lack of effective therapies targeting advanced fibrosis. Hydrogen peroxide-inducible clone-5 (Hic-5), an adaptor protein in focal adhesion, is critical for promoting liver fibrosis in hepatic stellate cells. This study investigated its clinical applicability by examining hepatic Hic-5 expression in human fibrotic tissues, exploring its association with MASH, and assessing the therapeutic potential of antisense oligonucleotides (ASOs) targeting Hic-5 in a MASH mouse model. Methods Hepatic Hic-5 expression in human fibrotic tissues underwent pathological image analysis and single-cell RNA sequencing. ASOs targeting Hic-5 were developed and tested using in vitro cell models. An in vivo MASH mouse model was used to evaluate the effects of anti-Hic-5 ASOs on advanced fibrosis and steatosis. Results Hepatic Hic-5 expression increased with the progression of fibrosis, particularly in advanced stages. Single-cell RNA sequencing revealed Hic-5 expression primarily in hepatic stellate cells. In MASH-associated fibrosis, Hic-5 expression correlated with the expression of fibrotic genes. In the MASH mouse model, hepatic Hic-5 expression increased with disease progression. Anti-Hic-5 ASOs effectively suppressed Hic-5 expression in vitro and attenuated advanced fibrosis and steatosis in vivo, indicating their therapeutic potential. Conclusions Hepatic Hic-5 expression is associated with advanced liver fibrosis and MASH. Anti-Hic-5 ASOs are promising therapeutic interventions for MASH accompanied by advanced fibrosis. These findings provide valuable insights into potential clinical treatments for advanced liver fibrosis. Impact and implications This study investigated the role of Hic-5 in liver fibrosis and steatohepatitis, highlighting its potential as a therapeutic target. We developed an antisense oligonucleotide (ASO) that was particularly transportable to the liver, and targeted Hic-5. Anti-Hic-5 ASO exhibited therapeutic efficacy for liver fibrosis and steatosis in vivo, indicating its therapeutic potential for liver fibrosis and steatosis. ASOs have already achieved dramatic therapeutic effects as approved nucleic acid drugs. Thus, anti-Hic-5 ASO is expected to lead the direct generation of seed compounds for the clinical development of drugs for liver fibrosis and steatosis.
Collapse
Affiliation(s)
- Masahito Noguchi
- Department of Biochemistry, Showa University School of Medicine; Shinagawa-ku, Tokyo, Japan
| | - Aya Miyauchi
- Department of Biochemistry, Showa University School of Medicine; Shinagawa-ku, Tokyo, Japan
- Institute for Extracellular Matrix Research, Showa University; Shinagawa-ku, Tokyo, Japan
| | - Yoshiaki Masaki
- Department of Life Science and Technology, Tokyo Institute of Technology; Yokohama, Kanagawa, Japan
| | - Masashi Sakaki
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine; Shinagawa-ku, Tokyo, Japan
| | - Xiao-Feng Lei
- Department of Dermatology, Showa University School of Medicine; Shinagawa-ku, Tokyo, Japan
| | | | - Akira Miyazaki
- Department of Biochemistry, Showa University School of Medicine; Shinagawa-ku, Tokyo, Japan
- Institute for Extracellular Matrix Research, Showa University; Shinagawa-ku, Tokyo, Japan
| | - Takeshi Aoki
- Department of General and Gastroenterological Surgery, Showa University School of Medicine; Shinagawa-ku, Tokyo, Japan
| | - Hitoshi Yoshida
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine; Shinagawa-ku, Tokyo, Japan
| | - Kohji Seio
- Department of Life Science and Technology, Tokyo Institute of Technology; Yokohama, Kanagawa, Japan
| | - Joo-ri Kim-Kaneyama
- Department of Biochemistry, Showa University School of Medicine; Shinagawa-ku, Tokyo, Japan
- Institute for Extracellular Matrix Research, Showa University; Shinagawa-ku, Tokyo, Japan
| |
Collapse
|
49
|
Agwa MM, Elmotasem H, El-Lakany SA. Small molecules and peptide ligands directed nano-therapeutics for precise oncological phototherapy: Emphasis towards enhancing chemotherapeutic active tumor targeting efficacy. J Drug Deliv Sci Technol 2024; 101:106313. [DOI: 10.1016/j.jddst.2024.106313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
50
|
Altinbay M, Wang J, Chen J, Schäfer D, Sprang M, Blagojevic B, Wölfl S, Andrade-Navarro M, Dikic I, Knapp S, Cheng X. Chem-CRISPR/dCas9FCPF: a platform for chemically induced epigenome editing. Nucleic Acids Res 2024; 52:11587-11601. [PMID: 39315698 PMCID: PMC11514490 DOI: 10.1093/nar/gkae798] [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: 06/04/2023] [Revised: 08/20/2024] [Accepted: 09/18/2024] [Indexed: 09/25/2024] Open
Abstract
Epigenetic aberration is one of the major driving factors in human cancer, often leading to acquired resistance to chemotherapies. Various small molecule epigenetic modulators have been reported. Nonetheless, outcomes from animal models and clinical trials have underscored the substantial setbacks attributed to pronounced on- and off-target toxicities. To address these challenges, CRISPR/dCas9 technology is emerging as a potent tool for precise modulation of epigenetic mechanism. However, this technology involves co-expressing exogenous epigenetic modulator proteins, which presents technical challenges in preparation and delivery with potential undesirable side effects. Recently, our research demonstrated that Cas9 tagged with the Phe-Cys-Pro-Phe (FCPF)-peptide motif can be specifically targeted by perfluorobiphenyl (PFB) derivatives. Here, we integrated the FCPF-tag into dCas9 and established a chemically inducible platform for epigenome editing, called Chem-CRISPR/dCas9FCPF. We designed a series of chemical inhibitor-PFB conjugates targeting various epigenetic modulator proteins. Focusing on JQ1, a panBET inhibitor, we demonstrate that c-MYC-sgRNA-guided JQ1-PFB specifically inhibits BRD4 in close proximity to the c-MYC promoter/enhancer, thereby effectively repressing the intricate transcription networks orchestrated by c-MYC as compared with JQ1 alone. In conclusion, our Chem-CRISPR/dCas9FCPF platform significantly increased target specificity of chemical epigenetic inhibitors, offering a viable alternative to conventional fusion protein systems for epigenome editing.
Collapse
Affiliation(s)
- Mukaddes Altinbay
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt am Main, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| | - Jianhui Wang
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt am Main, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| | - Jie Chen
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt am Main, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Paul-Ehrlich-Straße 42-44, 60596 Frankfurt am Main, Germany
- University Cancer Center (UCT) Frankfurt, Theodor-Stern-Kai7, 60590, Frankfurt am Main, Germany
| | - Daniel Schäfer
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt am Main, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| | - Maximilian Sprang
- Faculty of Biology, Johannes Gutenberg University Mainz, Gresemundweg 2, 55128 Mainz, Germany
| | - Biljana Blagojevic
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, Heidelberg, Germany
| | - Stefan Wölfl
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, Heidelberg, Germany
| | | | - Ivan Dikic
- Institute of Biochemistry II, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Stefan Knapp
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt am Main, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Paul-Ehrlich-Straße 42-44, 60596 Frankfurt am Main, Germany
- DKTK translational cancer network, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Xinlai Cheng
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt am Main, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Paul-Ehrlich-Straße 42-44, 60596 Frankfurt am Main, Germany
- University Cancer Center (UCT) Frankfurt, Theodor-Stern-Kai7, 60590, Frankfurt am Main, Germany
| |
Collapse
|