101
|
Lu X, Smaill JB, Ding K. Medicinal Chemistry Strategies for the Development of Kinase Inhibitors Targeting Point Mutations. J Med Chem 2020; 63:10726-10741. [PMID: 32432477 DOI: 10.1021/acs.jmedchem.0c00507] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Clinically acquired resistance to small molecule kinase inhibitors (SMKIs) has become a major "unmet clinical need" in cancer therapy. To date, there are six SMKIs to be approved for the treatment of cancer patients through targeting of clinically acquired resistance caused by on-target mutations. These are mainly focused on the mutant kinases Bcr-Abl T315I, EGFR T790M, and ALK L1196M. Herein, we summarize the major medicinal chemistry strategies employed in the discovery of these representative SMKIs, such as avoiding steric hindrance, making additional interactions with mutated residues, and forming a covalent bond with an active site cysteine to override resistance observed for reversible inhibitors. Additionally, we also briefly describe allosteric kinase inhibitors and proteolysis targeting chimera (PROTAC) as two other potential strategies while addressing future opportunities in this area.
Collapse
Affiliation(s)
- Xiaoyun Lu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Jeff B Smaill
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Ke Ding
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| |
Collapse
|
102
|
Hanan EJ, Liang J, Wang X, Blake RA, Blaquiere N, Staben ST. Monomeric Targeted Protein Degraders. J Med Chem 2020; 63:11330-11361. [DOI: 10.1021/acs.jmedchem.0c00093] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
103
|
Shu L, Chen C, Huan X, Huang H, Wang M, Zhang J, Yan Y, Liu J, Zhang T, Zhang D. Design, synthesis, and pharmacological evaluation of 4- or 6-phenyl-pyrimidine derivatives as novel and selective Janus kinase 3 inhibitors. Eur J Med Chem 2020; 191:112148. [PMID: 32097841 DOI: 10.1016/j.ejmech.2020.112148] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/13/2020] [Accepted: 02/13/2020] [Indexed: 12/11/2022]
Abstract
As non-receptor tyrosine kinases, Janus kinases (JAKs) have become an attractive target for the treatment of autoimmune diseases and cancers. JAKs play a pivotal role in innate immunity, inflammation, and hematopoiesis by mediating the signaling of numerous cytokines, growth factors, and interferons (IFNs). Selective inhibitors of a variety of JAK members are expected to inhibit pro-inflammatory cytokine-mediated inflammation and immune responses, while preventing targeting other subtypes of JAKs. In this work, poorly selective compounds based on 4- or 6-phenyl-pyrimidine derivatives have been improved to highly potent and selective compounds by designing a covalent binding tether, which attaches to the unique cysteine (Cys909) residue in JAK3. Compound 12 exhibited potent JAK3 inhibitory activity (IC50 = 1.7 nM) with an excellent selectivity profile when compared to the other JAK isoforms (>588-fold). In a cellular assay, compound 12 strongly inhibited JAK3-dependent signaling and T cell proliferation. Moreover, in vivo data revealed that compound 12 significantly suppressed oxazolone (OXZ)-induced delayed hypersensitivity responses in Balb/c mice. Compound 12 also displayed decent pharmacokinetic properties and was suitable for in vivo use. Taken together, these results indicated that compound 12 may be a promising tool compound as a selective JAK3 inhibitor for treating autoimmune diseases.
Collapse
Affiliation(s)
- Lei Shu
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Chengjuan Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100050, PR China
| | - Xueting Huan
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Hao Huang
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Manman Wang
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Jianqiu Zhang
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yile Yan
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Jianming Liu
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Tiantai Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100050, PR China.
| | - Dayong Zhang
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, PR China.
| |
Collapse
|
104
|
Ran F, Liu Y, Yu S, Guo K, Tang W, Chen X, Zhao G. Design and synthesis of novel 1-substituted 3-(6-phenoxypyridin-3-yl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine analogs as selective BTK inhibitors for the treatment of mantle cell lymphoma. Bioorg Chem 2020; 94:103367. [DOI: 10.1016/j.bioorg.2019.103367] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/16/2019] [Accepted: 10/14/2019] [Indexed: 11/26/2022]
|
105
|
Design, synthesis and biological evaluation of Proteolysis Targeting Chimeras (PROTACs) as a BTK degraders with improved pharmacokinetic properties. Bioorg Med Chem Lett 2019; 30:126877. [PMID: 31879210 DOI: 10.1016/j.bmcl.2019.126877] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 11/23/2022]
Abstract
A new series of Proteolysis Targeting Chimeras (PROTACs) targeting Bruton's Tyrosine Kinase (BTK) was synthesized, with the goal of improving the pharmacokinetic properties of our previously reported PROTAC, MT802. We recently described the ability of MT802 to induce degradation of both wild-type and C481S mutant BTK in immortalized cells and patient-derived B-lymphocytes. However, the pharmacokinetic properties of MT802 were not suitable for further in vivo development. Therefore, we undertook a systematic medicinal chemistry campaign to overcome this issue and made a series of PROTACs with structural modifications to the linker and E3-recruiting ligand; more specifically, the new PROTACs were synthesized with different von Hippel-Lindau (VHL) and cereblon (CRBN) ligands while keeping the BTK ligand and linker length constant. This approach resulted in an equally potent PROTAC, SJF620, with a significantly better pharmacokinetic profile than MT802. This compound may hold promise for further in vivo exploration of BTK degradation.
Collapse
|
106
|
Misselbeck K, Parolo S, Lorenzini F, Savoca V, Leonardelli L, Bora P, Morine MJ, Mione MC, Domenici E, Priami C. A network-based approach to identify deregulated pathways and drug effects in metabolic syndrome. Nat Commun 2019; 10:5215. [PMID: 31740673 PMCID: PMC6861239 DOI: 10.1038/s41467-019-13208-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/25/2019] [Indexed: 12/11/2022] Open
Abstract
Metabolic syndrome is a pathological condition characterized by obesity, hyperglycemia, hypertension, elevated levels of triglycerides and low levels of high-density lipoprotein cholesterol that increase cardiovascular disease risk and type 2 diabetes. Although numerous predisposing genetic risk factors have been identified, the biological mechanisms underlying this complex phenotype are not fully elucidated. Here we introduce a systems biology approach based on network analysis to investigate deregulated biological processes and subsequently identify drug repurposing candidates. A proximity score describing the interaction between drugs and pathways is defined by combining topological and functional similarities. The results of this computational framework highlight a prominent role of the immune system in metabolic syndrome and suggest a potential use of the BTK inhibitor ibrutinib as a novel pharmacological treatment. An experimental validation using a high fat diet-induced obesity model in zebrafish larvae shows the effectiveness of ibrutinib in lowering the inflammatory load due to macrophage accumulation.
Collapse
Affiliation(s)
- Karla Misselbeck
- Fondazione The Microsoft Research University of Trento, Centre for Computational and Systems Biology (COSBI), Rovereto, Italy
- Department of Mathematics, University of Trento, Trento, Italy
| | - Silvia Parolo
- Fondazione The Microsoft Research University of Trento, Centre for Computational and Systems Biology (COSBI), Rovereto, Italy.
| | - Francesca Lorenzini
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Valeria Savoca
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Lorena Leonardelli
- Fondazione The Microsoft Research University of Trento, Centre for Computational and Systems Biology (COSBI), Rovereto, Italy
| | - Pranami Bora
- Fondazione The Microsoft Research University of Trento, Centre for Computational and Systems Biology (COSBI), Rovereto, Italy
| | - Melissa J Morine
- Fondazione The Microsoft Research University of Trento, Centre for Computational and Systems Biology (COSBI), Rovereto, Italy
| | - Maria Caterina Mione
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Enrico Domenici
- Fondazione The Microsoft Research University of Trento, Centre for Computational and Systems Biology (COSBI), Rovereto, Italy.
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy.
| | - Corrado Priami
- Fondazione The Microsoft Research University of Trento, Centre for Computational and Systems Biology (COSBI), Rovereto, Italy.
- Department of Computer Science, University of Pisa, Pisa, Italy.
| |
Collapse
|
107
|
Xia LW, Ba MY, Liu W, Cheng W, Hu CP, Zhao Q, Yao YF, Sun MR, Duan YT. Triazol: a privileged scaffold for proteolysis targeting chimeras. Future Med Chem 2019; 11:2919-2973. [PMID: 31702389 DOI: 10.4155/fmc-2019-0159] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Current traditional drugs such as enzyme inhibitors and receptor agonists/antagonists present inherent limitations due to occupancy-driven pharmacology as the mode of action. Proteolysis targeting chimeras (PROTACs) are composed of an E3 ligand, a connecting linker and a target protein ligand, and are an attractive approach to specifically knockdown-targeted proteins utilizing an event-driven mode of action. The length, hydrophilicity and rigidity of connecting linkers play important role in creating a successful PROTAC. Some PROTACs with a triazole linker have displayed promising anticancer activity. This review provides an overview of PROTACs with a triazole scaffold and discusses its structure-activity relationship. Important milestones in the development of PROTACs are addressed and a critical analysis of this drug discovery strategy is also presented.
Collapse
Affiliation(s)
- Li-Wen Xia
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
- Collaborative Innovation Center of Henan New Drug Research & Safety Evaluation, Zhengzhou, Henan 450001, China
| | - Meng-Yu Ba
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
- Collaborative Innovation Center of Henan New Drug Research & Safety Evaluation, Zhengzhou, Henan 450001, China
| | - Wei Liu
- Henan Provincial Key Laboratory of Children's Genetics & Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou 450018, China
| | - Weyland Cheng
- Henan Provincial Key Laboratory of Children's Genetics & Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou 450018, China
| | - Chao-Ping Hu
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
- Collaborative Innovation Center of Henan New Drug Research & Safety Evaluation, Zhengzhou, Henan 450001, China
| | - Qing Zhao
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
- Collaborative Innovation Center of Henan New Drug Research & Safety Evaluation, Zhengzhou, Henan 450001, China
| | - Yong-Fang Yao
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
- Collaborative Innovation Center of Henan New Drug Research & Safety Evaluation, Zhengzhou, Henan 450001, China
| | - Mo-Ran Sun
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
- Collaborative Innovation Center of Henan New Drug Research & Safety Evaluation, Zhengzhou, Henan 450001, China
| | - Yong-Tao Duan
- Henan Provincial Key Laboratory of Children's Genetics & Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou 450018, China
| |
Collapse
|
108
|
New 2,6,9-trisubstituted purine derivatives as Bcr-Abl and Btk inhibitors and as promising agents against leukemia. Bioorg Chem 2019; 94:103361. [PMID: 31699386 DOI: 10.1016/j.bioorg.2019.103361] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 10/12/2019] [Indexed: 12/14/2022]
Abstract
Bcr-Abl and Btk kinases are among the targets that have been considered for the treatment of leukemia. Therefore, several strategies have focused on the use of inhibitors as chemotherapeutic tools to treat these types of leukemia, such as imatinib (for Bcr-Abl) or ibrutinib (for Btk). However, the efficacy of these drugs has been reduced due to resistance mechanisms, which have motivated the development of new and more effective compounds. In this study, we designed, synthesized and evaluated 2,6,9-trisubstituted purine derivatives as novel Bcr-Abl and Btk inhibitors. We identified 5c and 5d as potent inhibitors of both kinases (IC50 values of 40 nM and 0.58/0.66 μM for Abl and Btk, respectively). From docking and QSAR analyses, we concluded that fluorination of the arylpiperazine system is detrimental to the activity against two kinases, and we also validated our hypothesis that the substitution on the 6-phenylamino ring is important for the inhibition of both kinases. In addition, our studies indicated that most compounds could suppress the proliferation of leukemia and lymphoma cells (HL60, MV4-11, CEM, K562 and Ramos cells) at low micromolar concentrations in vitro. Finally, we preliminarily demonstrated that 5c inhibited the downstream signaling of both kinases in the respective cell models. Therefore, 5c or 5d possessed potency to be further optimized as anti-leukemia drugs by simultaneously inhibiting the Bcr-Abl and Btk kinases.
Collapse
|
109
|
Guo X, Yang D, Fan Z, Zhang N, Zhao B, Huang C, Wang F, Ma R, Meng M, Deng Y. Discovery and structure-activity relationship of novel diphenylthiazole derivatives as BTK inhibitor with potent activity against B cell lymphoma cell lines. Eur J Med Chem 2019; 178:767-781. [DOI: 10.1016/j.ejmech.2019.06.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 06/09/2019] [Accepted: 06/10/2019] [Indexed: 10/26/2022]
|
110
|
Abeykoon JP, Zanwar S, Ansell SM, Gertz MA, Kumar S, Manske M, Novak AJ, King R, Greipp P, Go R, Inwards D, Muchtar E, Habermann T, Witzig TE, Thompson CA, Dingli D, Lacy MQ, Leung N, Dispenzieri A, Gonsalves W, Warsame R, Kyle RA, Rajkumar V, Parikh SA, Kapoor P. Ibrutinib monotherapy outside of clinical trial setting in Waldenström macroglobulinaemia: practice patterns, toxicities and outcomes. Br J Haematol 2019; 188:394-403. [DOI: 10.1111/bjh.16168] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 06/28/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Jithma P. Abeykoon
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Saurabh Zanwar
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Stephen M. Ansell
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Morie A. Gertz
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Shaji Kumar
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Michelle Manske
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Anne J. Novak
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Rebecca King
- Department of Laboratory Medicine and Pathology Mayo Clinic Rochester MN USA
| | - Patricia Greipp
- Division of Laboratory Genetics Mayo Clinic Rochester MN USA
| | - Ronald Go
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - David Inwards
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Eli Muchtar
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Thomas Habermann
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Thomas E. Witzig
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Carrie A. Thompson
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - David Dingli
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Martha Q. Lacy
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Nelson Leung
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Angela Dispenzieri
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Wilson Gonsalves
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Rahma Warsame
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Robert A. Kyle
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Vincent Rajkumar
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Sameer A. Parikh
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Prashant Kapoor
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester MN USA
| |
Collapse
|
111
|
Abstract
Modification of pathogenic antibodies for autoimmune diseases illuminated the biologic relevance of B cells, plasma cells, and pathogenic antibodies in autoimmunity. They have also rejuvenated interest in how B cells mediate multiple effector functions that include antibody production, antigen presentation to T cells, costimulation, and the production of immune stimulating and immune modulatory cytokines. Repurposing these drugs from autoimmunity and cancer immunotherapy has yielded important advancements in the care of antibody-mediated rejection patients and novel drug development aimed at HLA desensitization have recently emerged. We now stand on an important threshold that promises many advances in the care of our allosensitized patients. We hope that these initial advances will encourage basic scientist, clinical investigators, industry, National Institutes of Health, our academic societies, and the Food and Drug Administration to continue support of these important objectives. These advances clearly have implications for sensitized patients receiving solid organ transplants and antibody-mediated rejection treatment. Modification of alloimmunity and alloantibodies will also have relevance to xenotransplantation where the xenoantibodies present a formidable obstacle to advancement of this important therapy. Working together, we can advance transplant therapeutics where biologic agents are likely to play novel and important roles. Here, we discuss novel drugs emerging in this area.
Collapse
|
112
|
Ran F, Liu Y, Liu M, Zhang D, Wang P, Dong J, Tang W, Zhao G. Discovery of pyrazolopyrimidine derivatives as potent BTK inhibitors with effective anticancer activity in MCL. Bioorg Chem 2019; 89:102943. [DOI: 10.1016/j.bioorg.2019.102943] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 04/03/2019] [Accepted: 04/19/2019] [Indexed: 02/04/2023]
|
113
|
Diao Y, Fang X, Song P, Lai M, Tong L, Hao Y, Dou D, Liu Y, Ding J, Zhao Z, Xie H, Li H. Discovery and Biological evaluation of pyrimido[4,5-d]pyrimidine-2,4(1H,3H)-dione derivatives as potent Bruton’s tyrosine kinase inhibitors. Bioorg Med Chem 2019; 27:3390-3395. [DOI: 10.1016/j.bmc.2019.06.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 12/14/2022]
|
114
|
Yao X, Sun X, Jin S, Yang L, Xu H, Rao Y. Discovery of 4-Aminoquinoline-3-carboxamide Derivatives as Potent Reversible Bruton’s Tyrosine Kinase Inhibitors for the Treatment of Rheumatoid Arthritis. J Med Chem 2019; 62:6561-6574. [DOI: 10.1021/acs.jmedchem.9b00329] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Xia Yao
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P. R. China
| | - Xiuyun Sun
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P. R. China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, P. R. China
| | - Shuyu Jin
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
| | - Ling Yang
- R&D Institute, Chia Tai Tianqing Pharmaceutical Group Co., LTD, Nanjing 210023, P. R. China
| | - Hongjiang Xu
- R&D Institute, Chia Tai Tianqing Pharmaceutical Group Co., LTD, Nanjing 210023, P. R. China
| | - Yu Rao
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
115
|
Giordano F, Vaira V, Cortinovis D, Bonomo S, Goedmakers J, Brena F, Cialdella A, Ianzano L, Forno I, Cerrito MG, Giovannoni R, Ferri GL, Tasciotti E, Vicent S, Damarco F, Bosari S, Lavitrano M, Grassilli E. p65BTK is a novel potential actionable target in KRAS-mutated/EGFR-wild type lung adenocarcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:260. [PMID: 31200752 PMCID: PMC6570906 DOI: 10.1186/s13046-019-1199-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Lung cancer is still the main cause of cancer death worldwide despite the availability of targeted therapies and immune-checkpoint inhibitors combined with chemotherapy. Cancer cell heterogeneity and primary or acquired resistance mechanisms cause the elusive behaviour of this cancer and new biomarkers and active drugs are urgently needed to overcome these limitations. p65BTK, a novel isoform of the Bruton Tyrosine Kinase may represent a new actionable target in non-small cell lung cancer (NSCLC). METHODS p65BTK expression was evaluated by immunohistochemistry in 382 NSCLC patients with complete clinico-pathological records including smoking habit, ALK and EGFR status, and in metastatic lymph nodes of 30 NSCLC patients. NSCLC cell lines mutated for p53 and/or a component of the RAS/MAPK pathway and primary lung cancer-derived cells from Kras/Trp53 null mice were used as a preclinical model. The effects of p65BTK inhibition by BTK Tyrosine Kinase Inhibitors (TKIs) (Ibrutinib, AVL-292, RN486) and first-generation EGFR-TKIs (Gefitinib, Erlotinib) on cell viability were evaluated by MTT. The effects of BTK-TKIs on cell growth and clonogenicity were assessed by crystal violet and colony assays, respectively. Cell toxicity assays were performed to study the effect of the combination of non-toxic concentrations of BTK-TKIs with EGFR-TKIs and standard-of-care (SOC) chemotherapy (Cisplatin, Gemcitabine, Pemetrexed). RESULTS p65BTK was significantly over-expressed in EGFR-wild type (wt) adenocarcinomas (AdC) from non-smoker patients and its expression was also preserved at the metastatic site. p65BTK was also over-expressed in cell lines mutated for KRAS or for a component of the RAS/MAPK pathway and in tumors from Kras/Trp53 null mice. BTK-TKIs were more effective than EGFR-TKIs in decreasing cancer cell viability and significantly impaired cell proliferation and clonogenicity. Moreover, non-toxic doses of BTK-TKIs re-sensitized drug-resistant NSCLC cell lines to both target- and SOC therapy, independently from EGFR/KRAS status. CONCLUSIONS p65BTK results as an emerging actionable target in non-smoking EGFR-wt AdC, also at advanced stages of disease. Notably, these patients are not eligible for EGFR-TKIs-based therapy due to a lack of EGFR mutation. The combination of BTK-TKIs with EGFR-TKIs is cytotoxic for EGFR-wt/KRAS-mutant/p53-null tumors and BTK-TKIs re-sensitizes drug-resistant NSCLC to SOC chemotherapy. Therefore, our data suggest that adding BTK-TKIs to SOC chemotherapy and EGFR-targeted therapy may open new avenues for clinical trials in currently untreatable NSCLC.
Collapse
Affiliation(s)
- Federica Giordano
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Valentina Vaira
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | | | - Sara Bonomo
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Joyce Goedmakers
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Federica Brena
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Annamaria Cialdella
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Leonarda Ianzano
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Irene Forno
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | | | - Roberto Giovannoni
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,Present address: Department of Biology, University of Pisa, Pisa, Italy
| | - Gian Luca Ferri
- Department of Biomedical Science, NEF-Laboratory, University of Cagliari, Cagliari, Italy
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, USA.,Houston Methodist Orthopedic and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Silve Vicent
- Center for Applied Medical Research, Program in Solid Tumors and Biomarkers, University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Francesco Damarco
- Division of Thoracic Surgery and Lung Tranplantation, Fondazione IRCCS Ca' Granda Ospedale maggiore Policlinico Milano, Milano, Italy
| | - Silvano Bosari
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | | | - Emanuela Grassilli
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.
| |
Collapse
|
116
|
Um IS, Armstrong-Gordon E, Moussa YE, Gnjidic D, Wheate NJ. Platinum drugs in the Australian cancer chemotherapy healthcare setting: Is it worthwhile for chemists to continue to develop platinums? Inorganica Chim Acta 2019. [DOI: 10.1016/j.ica.2019.04.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
117
|
Design, synthesis and evaluation of novel 7H-pyrrolo[2,3-d]pyrimidin-4-amine derivatives as potent, selective and reversible Bruton's tyrosine kinase (BTK) inhibitors for the treatment of rheumatoid arthritis. Eur J Med Chem 2019; 169:121-143. [DOI: 10.1016/j.ejmech.2019.02.077] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 01/29/2023]
|
118
|
Discovery of novel pyrazole derivatives as potential anticancer agents in MCL. Bioorg Med Chem Lett 2019; 29:1060-1064. [DOI: 10.1016/j.bmcl.2019.03.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 02/21/2019] [Accepted: 03/03/2019] [Indexed: 11/20/2022]
|
119
|
Feng Y, Duan W, Cu X, Liang C, Xin M. Bruton's tyrosine kinase (BTK) inhibitors in treating cancer: a patent review (2010-2018). Expert Opin Ther Pat 2019; 29:217-241. [PMID: 30888232 DOI: 10.1080/13543776.2019.1594777] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Bruton's tyrosine kinase (BTK) plays a critical role in the regulation of survival, proliferation, activation and differentiation of B-lineage cells. It participates by regulating multiple cellular signaling pathways, including B cell receptor and FcR signaling cascades. BTK is abundantly expressed and constitutively active in the pathogenesis of B cell hematological malignancies, as well as several autoimmune diseases. Therefore, BTK is considered as an attractive target for treatment of B-lineage lymphomas, leukemias, and some autoimmune diseases. Many industry and academia efforts have been made to explore small molecular BTK inhibitors. AREAS COVERED This review aims to provide an overview of the patented BTK inhibitors for the treatment of cancer from 2010 to 2018. EXPERT OPINION BTK inhibitors attract much interest for their therapeutic potential in the treatment of cancers and autoimmune diseases, especially for B cell hematological malignancies. In 2013, ibrutinib was approved by the FDA as the first-in-class BTK inhibitors for the treatment of mantle cell lymphoma (MCL) and chronic lymphocytic leukemia (CLL), and now it is also undergoing clinical evaluation for other indications in either single or combined therapy. It is clear that BTK inhibitors can provide a promising clinical benefit in treating B-lineage lymphomas and leukemias.
Collapse
Affiliation(s)
- Yifan Feng
- a Department of Medicinal Chemistry, School of Pharmacy, Health Science Center , Xi'an Jiaotong University , Xi'an , Shaanxi P.R. China
| | - Weiming Duan
- a Department of Medicinal Chemistry, School of Pharmacy, Health Science Center , Xi'an Jiaotong University , Xi'an , Shaanxi P.R. China
| | - Xiaochuan Cu
- b Department of Orthopedics , People's Hospital of Fufeng County in Shaanxi Province , Baoji , Shaanxi P.R. China
| | - Chengyuan Liang
- c Department of Pharmacy , Shaanxi University of Science & Technology , Xi'an , Shaanxi P.R. China
| | - Minhang Xin
- a Department of Medicinal Chemistry, School of Pharmacy, Health Science Center , Xi'an Jiaotong University , Xi'an , Shaanxi P.R. China
| |
Collapse
|
120
|
Are peptides a solution for the treatment of hyperactivated JAK3 pathways? Inflammopharmacology 2019; 27:433-452. [PMID: 30929155 DOI: 10.1007/s10787-019-00589-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/18/2019] [Indexed: 01/10/2023]
Abstract
While the inactivation mutations that eliminate JAK3 function lead to the immunological disorders such as severe combined immunodeficiency, activation mutations, causing constitutive JAK3 signaling, are known to trigger various types of cancer or are responsible for autoimmune diseases, such as rheumatoid arthritis, psoriasis, or inflammatory bowel diseases. Treatment of hyperactivated JAK3 is still an obstacle, due to different sensibility of mutation types to conventional drugs and unwanted side effects, because these drugs are not absolutely specific for JAK3, thus inhibiting other members of the JAK family, too. Lack of information, in which way sole inhibition of JAK3 is necessary for elimination of the disease, calls for the development of isoform-specific JAK3 inhibitors. Beside this strategy, up to date peptides are a rising alternative as chemo- or immunotherapeutics, but still sparsely represented in drug development and clinical trials. Beyond a possible direct inhibition function, crossing the cancer cell membrane and interfering in disease-causing pathways or triggering apoptosis, peptides could be used in future as adjunct remedies to potentialize traditional therapy and preserve non-affected cells. To discuss such feasible topics, this review deals with the knowledge about the structure-function of JAK3 and the actual state-of-the-art of isoform-specific inhibitor development, as well as the function of currently approved drugs or those currently being tested in clinical trials. Furthermore, several strategies for the application of peptide-based drugs for cancer therapy and the physicochemical and structural relations to peptide efficacy are discussed, and an overview of peptide sequences, which were qualified for clinical trials, is given.
Collapse
|
121
|
Postactivated B cells in systemic lupus erythematosus: update on translational aspects and therapeutic considerations. Curr Opin Rheumatol 2019; 31:175-184. [DOI: 10.1097/bor.0000000000000576] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
122
|
Li X, Shi B, Teng Y, Cheng Y, Yang H, Li J, Wang L, He S, You Q, Xiang H. Design, synthesis and biological evaluation of novel 2-phenyl pyrimidine derivatives as potent Bruton's tyrosine kinase (BTK) inhibitors. MEDCHEMCOMM 2019; 10:294-299. [PMID: 30881616 DOI: 10.1039/c8md00413g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/23/2018] [Indexed: 12/27/2022]
Abstract
BTK is an effective target for the treatment of B-cell malignant tumors and autoimmune diseases. In this work, a series of 2-phenyl pyrimidine derivatives were prepared and their preliminary in vitro activities on B-cell leukemia cells as well as the BTK enzyme were determined. The results showed that compound 11g displayed the best inhibitory activity on BTK with an inhibition rate of 82.76% at 100 nM and excellent anti-proliferation activity on three B-cell leukemia lines (IC50 = 3.66 μM, 6.98 μM, and 5.39 μM against HL60, Raji and Ramos, respectively). Besides, the flow cytometry analysis results indicated that 11g inhibited the proliferation of the Raji cells in a dose- and time-dependent manner, and blocked the Ramos cells at the G0/G1 phase, which is in accordance with the positive control ibrutinib. The mechanism investigation demonstrated that 11g could inhibit the phosphorylation of BTK and its downstream substrate phospholipase γ2 (PLCγ2). All these results showed that 11g was a promising lead compound that merited further optimization as a novel class of BTK inhibitor for the treatment of B-cell lymphoblastic leukemia.
Collapse
Affiliation(s)
- Xinyu Li
- Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China . ; ; Tel: +86 025 83271096
| | - Binyu Shi
- Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China . ; ; Tel: +86 025 83271096
| | - Yu Teng
- Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China . ; ; Tel: +86 025 83271096
| | - Yu Cheng
- Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China . ; ; Tel: +86 025 83271096
| | - Huizhu Yang
- Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China . ; ; Tel: +86 025 83271096
| | - Jiurong Li
- Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China . ; ; Tel: +86 025 83271096
| | - Lianjian Wang
- Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China . ; ; Tel: +86 025 83271096
| | - Siying He
- Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China . ; ; Tel: +86 025 83271096
| | - Qidong You
- Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China . ; ; Tel: +86 025 83271096
| | - Hua Xiang
- Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 210009 , China . ; ; Tel: +86 025 83271096
| |
Collapse
|
123
|
Kawahata W, Asami T, Kiyoi T, Irie T, Taniguchi H, Asamitsu Y, Inoue T, Miyake T, Sawa M. Design and Synthesis of Novel Amino-triazine Analogues as Selective Bruton's Tyrosine Kinase Inhibitors for Treatment of Rheumatoid Arthritis. J Med Chem 2018; 61:8917-8933. [PMID: 30216722 DOI: 10.1021/acs.jmedchem.8b01147] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Bruton's tyrosine kinase (BTK) is a promising drug target for the treatment of multiple diseases, such as B-cell malignances, asthma, and rheumatoid arthritis. A series of novel aminotriazines were identified as highly selective inhibitors of BTK by a scaffold-hopping approach. Subsequent SAR studies of this series using two conformationally different BTK proteins, an activated form of BTK and an unactivated form of BTK, led to the discovery of a highly selective BTK inhibitor, 4b. With significant efficacy in models in vivo and good ADME and safety profiles, 4b was advanced into preclinical studies.
Collapse
Affiliation(s)
- Wataru Kawahata
- Research and Development , Carna Biosciences, Inc. , 3rd Floor, BMA, 1-5-5 Minatojima-Minamimachi , Chuo-ku, Kobe 650-0047 , Japan
| | - Tokiko Asami
- Research and Development , Carna Biosciences, Inc. , 3rd Floor, BMA, 1-5-5 Minatojima-Minamimachi , Chuo-ku, Kobe 650-0047 , Japan
| | - Takao Kiyoi
- Research and Development , Carna Biosciences, Inc. , 3rd Floor, BMA, 1-5-5 Minatojima-Minamimachi , Chuo-ku, Kobe 650-0047 , Japan
| | - Takayuki Irie
- Research and Development , Carna Biosciences, Inc. , 3rd Floor, BMA, 1-5-5 Minatojima-Minamimachi , Chuo-ku, Kobe 650-0047 , Japan
| | - Haruka Taniguchi
- Research and Development , Carna Biosciences, Inc. , 3rd Floor, BMA, 1-5-5 Minatojima-Minamimachi , Chuo-ku, Kobe 650-0047 , Japan
| | - Yuko Asamitsu
- Research and Development , Carna Biosciences, Inc. , 3rd Floor, BMA, 1-5-5 Minatojima-Minamimachi , Chuo-ku, Kobe 650-0047 , Japan
| | - Tomoko Inoue
- Research and Development , Carna Biosciences, Inc. , 3rd Floor, BMA, 1-5-5 Minatojima-Minamimachi , Chuo-ku, Kobe 650-0047 , Japan
| | - Takahiro Miyake
- Research and Development , Carna Biosciences, Inc. , 3rd Floor, BMA, 1-5-5 Minatojima-Minamimachi , Chuo-ku, Kobe 650-0047 , Japan
| | - Masaaki Sawa
- Research and Development , Carna Biosciences, Inc. , 3rd Floor, BMA, 1-5-5 Minatojima-Minamimachi , Chuo-ku, Kobe 650-0047 , Japan
| |
Collapse
|
124
|
Li Z, Zhang L, Yuan Y, Yang Z. Identification of metabolites of evobrutinib in rat and human hepatocytes by using ultra-high performance liquid chromatography coupled with diode array detector and Q Exactive Orbitrap tandem mass spectrometry. Drug Test Anal 2018; 11:129-139. [PMID: 30102849 DOI: 10.1002/dta.2477] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/20/2018] [Accepted: 08/02/2018] [Indexed: 01/19/2023]
Affiliation(s)
- Zeyun Li
- Department of Pharmacy; the First Affiliated Hospital of Zhengzhou University; Zhengzhou China
| | - Lizhen Zhang
- Department of Pharmacy; the First Affiliated Hospital of Zhengzhou University; Zhengzhou China
| | - Yongliang Yuan
- Department of Pharmacy; the First Affiliated Hospital of Zhengzhou University; Zhengzhou China
| | - Zhiheng Yang
- Department of Pharmacy; the First Affiliated Hospital of Zhengzhou University; Zhengzhou China
| |
Collapse
|