1
|
Gong B, Liu Y, Li H, Ju X, Li D, Zou Y, Guo X, Dong K, Xiao J, Wu W, Chai R, Zhang R, Yu M. A Silk Fibroin Nanoparticle Hydrogel Loaded With NK1R Antagonist Has Synergistic Anti-Inflammatory and Reparative Effects on Dry Eye Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404835. [PMID: 39985258 PMCID: PMC12005769 DOI: 10.1002/advs.202404835] [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: 05/05/2024] [Revised: 01/28/2025] [Indexed: 02/24/2025]
Abstract
Dry eye disease (DED) is a multifactorial illness affecting tears and the ocular surface. The neurokinin 1 receptor (NK1R) is a target for controlling T helper 17 (Th17) and regulatory T cell (Treg) imbalances. This work creates a silk fibroin (SF) nanoparticle hydrogel that targets NK1R with CP-99,994 (CP). Combining CP and SF to generate stable nanoparticles while integrating a flexible hydrogel material results in a sustained-release ophthalmic drop formulation (SF@CP@Gel), which provides a long-lasting ocular formulation with anti-inflammatory and reparative properties. SF@CP@Gel could maintain a stable CP concentration for 25 h with detectable biological activity. The cell counting kit-8 and 2,7-DHL-DA results reveal that SF@CP@Gel has no cytotoxic effect on human corneal epithelial cells (HCECs) and decreases the reactive oxygen species level in oxidatively damaged HCECs. Cell scratch assays demonstrate that SF@CP@Gel can greatly increase HCEC migration and proliferation within 24 h. Furthermore, in vivo therapy with topical SF@CP@Gel twice daily markedly reduce clinical symptoms by reducing the amount of pathogenic Th17 cells while efficiently restoring Treg activity. In summary, this work reveals that SF@CP@Gel might attenuate DED by inhibiting NK1R-mediated SP signaling and thereby modulating the Th17/Treg ratio, a potential anti-inflammatory and repair treatment method for DED.
Collapse
Affiliation(s)
- Bo Gong
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- Human Disease Genes Key Laboratory of Sichuan Province and Institute of Laboratory MedicineSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Yi Liu
- Department of OphthalmologyDeyang People's HospitalDeyang618000China
- School of MedicineUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Huan Li
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- Department of OphthalmologyDeyang People's HospitalDeyang618000China
| | - Xueming Ju
- Human Disease Genes Key Laboratory of Sichuan Province and Institute of Laboratory MedicineSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Dongfeng Li
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Yuhao Zou
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Xiaoxin Guo
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- Human Disease Genes Key Laboratory of Sichuan Province and Institute of Laboratory MedicineSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Kai Dong
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- Human Disease Genes Key Laboratory of Sichuan Province and Institute of Laboratory MedicineSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Jialing Xiao
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- School of MedicineUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Weijia Wu
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- School of MedicineUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology‐Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologySchool of Medicine, Advanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjing210096China
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantong226001China
- Department of NeurologyAerospace Center HospitalSchool of Life ScienceBeijing Institute of TechnologyBeijing100081China
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaChengdu610072China
- Southeast University Shenzhen Research InstituteShenzhen518063China
| | - Ruifan Zhang
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Man Yu
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| |
Collapse
|
2
|
Kos J, Langiu M, Hellyer SD, Gregory KJ. Pharmacology, Signaling and Therapeutic Potential of Metabotropic Glutamate Receptor 5 Negative Allosteric Modulators. ACS Pharmacol Transl Sci 2024; 7:3671-3690. [PMID: 39698283 PMCID: PMC11651194 DOI: 10.1021/acsptsci.4c00213] [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: 04/11/2024] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 12/20/2024]
Abstract
Metabotropic glutamate receptors are a family of eight class C G protein-coupled receptors regulating higher order brain functions including cognition and motion. Metabotropic glutamate receptors have thus been heavily investigated as potential drug targets for treating neurological disorders. Drug discovery efforts directed toward metabotropic glutamate receptor subtype 5 (mGlu5) have been particularly fruitful, with a wealth of drug candidates and pharmacological tools identified. mGlu5 negative allosteric modulators (NAMs) are promising novel therapeutics for developmental, neuropsychiatric and neurodegenerative disorders (e.g., Alzheimer's Disease, Huntington's Disease, Parkinson's Disease, amyotrophic lateral sclerosis, autism spectrum disorders, substance use disorders, stroke, anxiety and depression) and show promise in ameliorating adverse effects induced by other medications (e.g., L-dopa induced dyskinesia in Parkinson's Disease). However, despite preclinical success, mGlu5 NAMs are yet to reach the market due to poor safety and efficacy profiles in clinical trials. Herein, we review the physiology and signal transduction of mGlu5. We provide a comprehensive critique of therapeutic options with respect to mGlu5 inhibitors, spanning from orthosteric antagonists to NAMs. Finally, we address the challenges associated with drug development and highlight future directions to guide rational drug discovery of safe and effective novel therapeutics.
Collapse
Affiliation(s)
- Jackson
A. Kos
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
| | - Monica Langiu
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
| | - Shane D. Hellyer
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
| | - Karen J. Gregory
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
- ARC
Centre for Cryo-electron Microscopy of Membrane Proteins, Monash University, Parkville, VIC 3052, Australia
| |
Collapse
|
3
|
Guo HW, Ye ZM, Chen SQ, McElwee KJ. Innovative strategies for the discovery of new drugs against alopecia areata: taking aim at the immune system. Expert Opin Drug Discov 2024; 19:1321-1338. [PMID: 39360759 DOI: 10.1080/17460441.2024.2409660] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024]
Abstract
INTRODUCTION The autoimmune hair loss condition alopecia areata (AA) exacts a substantial psychological and socioeconomic toll on patients. Biotechnology companies, dermatology clinics, and research institutions are dedicated to understanding AA pathogenesis and developing new therapeutic approaches. Despite recent efforts, many knowledge gaps persist, and multiple treatment development avenues remain unexplored. AREAS COVERED This review summarizes key AA disease mechanisms, current therapeutic methods, and emerging treatments, including Janus Kinase (JAK) inhibitors. The authors determine that innovative drug discovery strategies for AA are still needed due to continued unmet medical needs and the limited efficacy of current and emerging therapeutics. For prospective AA treatment developers, the authors identify the pre-clinical disease models available, their advantages, and limitations. Further, they outline treatment development opportunities that remain largely unmapped. EXPERT OPINION While recent advancements in AA therapeutics are promising, challenges remain, including the lack of consistent treatment efficacy, long-term use and safety issues, drug costs, and patient compliance. Future drug development research should focus on patient stratification utilizing robust biomarkers of AA disease activity and improved quantification of treatment response. Investigating superior modes of drug application and developing combination therapies may further improve outcomes. Spirited innovation will be needed to advance more effective treatments for AA.
Collapse
Affiliation(s)
- Hong-Wei Guo
- Department of Dermatology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhi-Ming Ye
- Guangdong Medical University, Zhanjiang, China
| | - Si-Qi Chen
- Guangdong Medical University, Zhanjiang, China
| | - Kevin J McElwee
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, Canada
- Centre for Skin Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
4
|
Rutkowski D, Scholey R, Davies J, Pye D, Blackhall F, Warren RB, Jimenez F, Griffiths CEM, Paus R. Epidermal growth factor receptor/mitogen-activated kinase inhibitor treatment induces a distinct inflammatory hair follicle response that includes collapse of immune privilege. Br J Dermatol 2024; 191:791-804. [PMID: 38857906 DOI: 10.1093/bjd/ljae243] [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: 01/26/2024] [Revised: 05/10/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Inhibitors of epidermal growth factor receptor (EGFRi) or mitogen-activated kinase (MEKi) induce a folliculitis in 75-90% of patients, the pathobiology of which remains insufficiently understood. OBJECTIVES To characterize changes in the skin immune status and global transcriptional profile of patients treated with EGFRi; to investigate whether EGFRi affects the hair follicle's (HF) immune privilege (IP); and to identify early proinflammatory signals induced by EGFRi/MEKi in human scalp HFs ex vivo. METHODS Scalp biopsies were taken from patients exhibiting folliculitis treated long term with EGFRi ('chronic EGFRi' group, n = 9) vs. healthy scalp skin (n = 9) and patients prior to commencing EGFRi treatment and after 2 weeks of EGFRi therapy ('acute EGFRi' group, n = 5). Healthy organ-cultured scalp HFs were exposed to an EGFRi (erlotinib, n = 5) or a MEKi (cobimetinib, n = 5). Samples were assessed by quantitative immunohistomorphometry, RNA sequencing (RNAseq) and in situ hybridization. RESULTS The 'chronic EGFRi' group showed CD8+ T-cell infiltration of the bulge alongside a partial collapse of the HF's IP, evidenced by upregulated major histocompatibility complex (MHC) class I, β2-microglobulin (B2 M) and MHC class II, and decreased transforming growth factor-β1 protein expression. Healthy HFs treated with EGFRi/MEKi ex vivo also showed partial HF IP collapse and increased transcription of human leucocyte antigen (HLA)-A, HLA-DR and B2 M transcripts. RNAseq analysis showed increased transcription of chemokines (CXCL1, CXCL13, CCL18, CCL3, CCL7) and interleukin (IL)-26 in biopsies from the 'chronic EGFRi' cohort, as well as increased IL-33 and decreased IL-37 expression in HF biopsies from the 'acute EGFRi' group and in organ-cultured HFs. CONCLUSIONS The data show that EGFRi/MEKi compromise the physiological IP of human scalp HFs and suggest that future clinical management of EGFRi/MEKi-induced folliculitis requires HF IP protection and inhibition of IL-33.
Collapse
Affiliation(s)
- David Rutkowski
- Dermatology Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, Manchester, UK
- Manchester University Foundation Trust, Manchester, UK
| | | | - John Davies
- Department of Safety Assessment, Genentech, Inc., South San Francisco, CA, USA
| | - Derek Pye
- Dermatology Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, Manchester, UK
| | | | - Richard B Warren
- Dermatology Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, Manchester, UK
| | - Francisco Jimenez
- Mediteknia Skin and Hair Lab, Las Palmas de Gran Canaria, Spain
- Universidad Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Christopher E M Griffiths
- Dermatology Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, Manchester, UK
- Department of Dermatology, King's College Hospital, King's College London, London, UK
| | - Ralf Paus
- Dermatology Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, Manchester, UK
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Monasterium Laboratory, Münster, Germany
- CUTANEON - Skin & Hair Innovations, Hamburg, Germany
| |
Collapse
|
5
|
Ohno Y, Suzuki M, Asada H, Kanda T, Saki M, Miyagi H, Yasunaga M, Suno C, Iwata S, Saito JI, Uchida S. In Vitro Pharmacological Profile of KW-6356, a Novel Adenosine A 2A Receptor Antagonist/Inverse Agonist. Mol Pharmacol 2023; 103:311-324. [PMID: 36894319 DOI: 10.1124/molpharm.122.000633] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 03/11/2023] Open
Abstract
KW-6356 is a novel adenosine A2A (A2A) receptor antagonist/inverse agonist, and its efficacy as monotherapy in Parkinson's disease (PD) patients has been reported. Istradefylline is a first-generation A2A receptor antagonist approved for use as adjunct treatment to levodopa/decarboxylase inhibitor in adult PD patients experiencing "OFF" episodes. In this study, we investigated the in vitro pharmacological profile of KW-6356 as an A2A receptor antagonist/inverse agonist and the mode of antagonism and compared them with istradefylline. In addition, we determined cocrystal structures of A2A receptor in complex with KW-6356 and istradefylline to explore the structural basis of the antagonistic properties of KW-6356. Pharmacological studies have shown that KW-6356 is a potent and selective ligand for the A2A receptor (the -log of inhibition constant = 9.93 ± 0.01 for human receptor) with a very low dissociation rate from the receptor (the dissociation kinetic rate constant = 0.016 ± 0.006 minute-1 for human receptor). In particular, in vitro functional studies indicated that KW-6356 exhibits insurmountable antagonism and inverse agonism, whereas istradefylline exhibits surmountable antagonism. Crystallography of KW-6356- and istradefylline-bound A2A receptor have indicated that interactions with His2506.52 and Trp2466.48 are essential for the inverse agonism, whereas the interactions at both deep inside the orthosteric pocket and the pocket lid stabilizing the extracellular loop conformation may contribute to the insurmountable antagonism of KW-6356. These profiles may reflect important differences in vivo and help predict better clinical performance. SIGNIFICANCE STATEMENT: KW-6356 is a potent and selective adenosine A2A receptor antagonist/inverse agonist and exhibits insurmountable antagonism, whereas istradefylline, a first-generation adenosine A2A receptor antagonist, exhibits surmountable antagonism. Structural studies of adenosine A2A receptor in complex with KW-6356 and istradefylline explain the characteristic differences in the pharmacological properties of KW-6356 and istradefylline.
Collapse
Affiliation(s)
- Yutaro Ohno
- Biomedical Science Research Laboratories 1 (Y.O., S.U.) and Molecular Analysis Center (Mi.S., H.M., J.S.), Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan; Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.A., S.I.); R&D Planning Department, R&D Division, Kyowa Kirin Co., Ltd., Tokyo, Japan (T.K.); Medical Affairs Department, Kyowa Kirin Co., Ltd., Tokyo, Japan (Ma.S.); CMC R&D Center, Production Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan (M.Y.); and Department of Medical Chemistry, Kansai Medical University, Osaka, Japan (C.S.)
| | - Michihiko Suzuki
- Biomedical Science Research Laboratories 1 (Y.O., S.U.) and Molecular Analysis Center (Mi.S., H.M., J.S.), Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan; Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.A., S.I.); R&D Planning Department, R&D Division, Kyowa Kirin Co., Ltd., Tokyo, Japan (T.K.); Medical Affairs Department, Kyowa Kirin Co., Ltd., Tokyo, Japan (Ma.S.); CMC R&D Center, Production Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan (M.Y.); and Department of Medical Chemistry, Kansai Medical University, Osaka, Japan (C.S.)
| | - Hidetsugu Asada
- Biomedical Science Research Laboratories 1 (Y.O., S.U.) and Molecular Analysis Center (Mi.S., H.M., J.S.), Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan; Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.A., S.I.); R&D Planning Department, R&D Division, Kyowa Kirin Co., Ltd., Tokyo, Japan (T.K.); Medical Affairs Department, Kyowa Kirin Co., Ltd., Tokyo, Japan (Ma.S.); CMC R&D Center, Production Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan (M.Y.); and Department of Medical Chemistry, Kansai Medical University, Osaka, Japan (C.S.)
| | - Tomoyuki Kanda
- Biomedical Science Research Laboratories 1 (Y.O., S.U.) and Molecular Analysis Center (Mi.S., H.M., J.S.), Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan; Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.A., S.I.); R&D Planning Department, R&D Division, Kyowa Kirin Co., Ltd., Tokyo, Japan (T.K.); Medical Affairs Department, Kyowa Kirin Co., Ltd., Tokyo, Japan (Ma.S.); CMC R&D Center, Production Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan (M.Y.); and Department of Medical Chemistry, Kansai Medical University, Osaka, Japan (C.S.)
| | - Mayumi Saki
- Biomedical Science Research Laboratories 1 (Y.O., S.U.) and Molecular Analysis Center (Mi.S., H.M., J.S.), Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan; Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.A., S.I.); R&D Planning Department, R&D Division, Kyowa Kirin Co., Ltd., Tokyo, Japan (T.K.); Medical Affairs Department, Kyowa Kirin Co., Ltd., Tokyo, Japan (Ma.S.); CMC R&D Center, Production Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan (M.Y.); and Department of Medical Chemistry, Kansai Medical University, Osaka, Japan (C.S.)
| | - Hikaru Miyagi
- Biomedical Science Research Laboratories 1 (Y.O., S.U.) and Molecular Analysis Center (Mi.S., H.M., J.S.), Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan; Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.A., S.I.); R&D Planning Department, R&D Division, Kyowa Kirin Co., Ltd., Tokyo, Japan (T.K.); Medical Affairs Department, Kyowa Kirin Co., Ltd., Tokyo, Japan (Ma.S.); CMC R&D Center, Production Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan (M.Y.); and Department of Medical Chemistry, Kansai Medical University, Osaka, Japan (C.S.)
| | - Mai Yasunaga
- Biomedical Science Research Laboratories 1 (Y.O., S.U.) and Molecular Analysis Center (Mi.S., H.M., J.S.), Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan; Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.A., S.I.); R&D Planning Department, R&D Division, Kyowa Kirin Co., Ltd., Tokyo, Japan (T.K.); Medical Affairs Department, Kyowa Kirin Co., Ltd., Tokyo, Japan (Ma.S.); CMC R&D Center, Production Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan (M.Y.); and Department of Medical Chemistry, Kansai Medical University, Osaka, Japan (C.S.)
| | - Chiyo Suno
- Biomedical Science Research Laboratories 1 (Y.O., S.U.) and Molecular Analysis Center (Mi.S., H.M., J.S.), Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan; Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.A., S.I.); R&D Planning Department, R&D Division, Kyowa Kirin Co., Ltd., Tokyo, Japan (T.K.); Medical Affairs Department, Kyowa Kirin Co., Ltd., Tokyo, Japan (Ma.S.); CMC R&D Center, Production Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan (M.Y.); and Department of Medical Chemistry, Kansai Medical University, Osaka, Japan (C.S.)
| | - So Iwata
- Biomedical Science Research Laboratories 1 (Y.O., S.U.) and Molecular Analysis Center (Mi.S., H.M., J.S.), Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan; Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.A., S.I.); R&D Planning Department, R&D Division, Kyowa Kirin Co., Ltd., Tokyo, Japan (T.K.); Medical Affairs Department, Kyowa Kirin Co., Ltd., Tokyo, Japan (Ma.S.); CMC R&D Center, Production Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan (M.Y.); and Department of Medical Chemistry, Kansai Medical University, Osaka, Japan (C.S.)
| | - Jun-Ichi Saito
- Biomedical Science Research Laboratories 1 (Y.O., S.U.) and Molecular Analysis Center (Mi.S., H.M., J.S.), Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan; Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.A., S.I.); R&D Planning Department, R&D Division, Kyowa Kirin Co., Ltd., Tokyo, Japan (T.K.); Medical Affairs Department, Kyowa Kirin Co., Ltd., Tokyo, Japan (Ma.S.); CMC R&D Center, Production Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan (M.Y.); and Department of Medical Chemistry, Kansai Medical University, Osaka, Japan (C.S.)
| | - Shinichi Uchida
- Biomedical Science Research Laboratories 1 (Y.O., S.U.) and Molecular Analysis Center (Mi.S., H.M., J.S.), Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan; Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.A., S.I.); R&D Planning Department, R&D Division, Kyowa Kirin Co., Ltd., Tokyo, Japan (T.K.); Medical Affairs Department, Kyowa Kirin Co., Ltd., Tokyo, Japan (Ma.S.); CMC R&D Center, Production Division, Kyowa Kirin Co., Ltd., Shizuoka, Japan (M.Y.); and Department of Medical Chemistry, Kansai Medical University, Osaka, Japan (C.S.)
| |
Collapse
|
6
|
Ohno Y, Okita E, Kawai-Uchida M, Fukuda N, Shoukei Y, Soshiroda K, Yamada K, Kanda T, Uchida S. Anti-parkinsonian activity of the adenosine A 2A receptor antagonist/inverse agonist KW-6356 as monotherapy in MPTP-treated common marmosets. Eur J Pharmacol 2023; 950:175773. [PMID: 37146707 DOI: 10.1016/j.ejphar.2023.175773] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/07/2023]
Abstract
KW-6356 is a novel adenosine A2A receptor antagonist/inverse agonist that not only blocks binding of adenosine to adenosine A2A receptor but also inhibits the constitutive activity of adenosine A2A receptor. The efficacy of KW-6356 as both monotherapy and an adjunct therapy to L-3,4-dihydroxyphenylalanine (L-DOPA)/decarboxylase inhibitor in Parkinson's disease (PD) patients has been reported. However, the first-generation A2A antagonist istradefylline, which is approved for use as an adjunct treatment to L-DOPA/decarboxylase inhibitor in adult PD patients experiencing OFF episodes, has not shown statistically significant efficacy as monotherapy. In vitro pharmacological studies have shown that the pharmacological properties of KW-6356 and istradefylline at adenosine A2A receptor are markedly different. However, the anti-parkinsonian activity and effects on dyskinesia of KW-6356 in PD animal models and the differences in the efficacy between KW-6356 and istradefylline are unknown. The present study investigated the anti-parkinsonian activity of KW-6356 as monotherapy in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated common marmosets, and its efficacy was directly compared with that of istradefylline. In addition, we investigated whether or not repeated administration of KW-6356 induced dyskinesia. Oral administration of KW-6356 reversed motor disability in a dose-dependent manner up to 1 mg/kg in MPTP-treated common marmosets. The magnitude of anti-parkinsonian activity induced by KW-6356 was significantly greater than that of istradefylline. Repeated administration of KW-6356 induced little dyskinesia in MPTP-treated common marmosets primed to exhibit dyskinesia by prior exposure to L-DOPA. These results indicate that KW-6356 can be a novel non-dopaminergic therapy as monotherapy without inducing dyskinesia in PD patients.
Collapse
Affiliation(s)
- Yutaro Ohno
- Biomedical Science Research Laboratories 1, Research Unit, R&D Division, Kyowa Kirin Co., Ltd., 1188 Shimotogari, Nagaizumi-cho, Sunto-gun, Shizuoka, 411-8731, Japan
| | - Eri Okita
- Research Core Function Laboratories, Research Unit, R&D Division, Kyowa Kirin Co., Ltd., 1188 Shimotogari, Nagaizumi-cho, Sunto-gun, Shizuoka, 411-8731, Japan
| | - Mika Kawai-Uchida
- Research Core Function Laboratories, Research Unit, R&D Division, Kyowa Kirin Co., Ltd., 1188 Shimotogari, Nagaizumi-cho, Sunto-gun, Shizuoka, 411-8731, Japan
| | - Naoko Fukuda
- Research Core Function Laboratories, Research Unit, R&D Division, Kyowa Kirin Co., Ltd., 1188 Shimotogari, Nagaizumi-cho, Sunto-gun, Shizuoka, 411-8731, Japan
| | - Youji Shoukei
- Research Core Function Laboratories, Research Unit, R&D Division, Kyowa Kirin Co., Ltd., 1188 Shimotogari, Nagaizumi-cho, Sunto-gun, Shizuoka, 411-8731, Japan
| | - Kazuhiro Soshiroda
- Research Core Function Laboratories, Research Unit, R&D Division, Kyowa Kirin Co., Ltd., 3-6-6 Asahi-machi, Machida-shi, Tokyo, 194-8533, Japan
| | - Koji Yamada
- Research Core Function Laboratories, Research Unit, R&D Division, Kyowa Kirin Co., Ltd., 1188 Shimotogari, Nagaizumi-cho, Sunto-gun, Shizuoka, 411-8731, Japan
| | - Tomoyuki Kanda
- R&D Planning Department, R&D Division, Kyowa Kirin Co., Ltd., 1-9-2 Otemachi, Chiyoda-ku, Tokyo, 100-0004, Japan
| | - Shinichi Uchida
- Biomedical Science Research Laboratories 1, Research Unit, R&D Division, Kyowa Kirin Co., Ltd., 1188 Shimotogari, Nagaizumi-cho, Sunto-gun, Shizuoka, 411-8731, Japan.
| |
Collapse
|
7
|
Vlachodimou A, de Vries H, Pasoli M, Goudswaard M, Kim SA, Kim YC, Scortichini M, Marshall M, Linden J, Heitman LH, Jacobson KA, IJzerman AP. Kinetic profiling and functional characterization of 8-phenylxanthine derivatives as A 2B adenosine receptor antagonists. Biochem Pharmacol 2022; 200:115027. [PMID: 35395239 DOI: 10.1016/j.bcp.2022.115027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 12/30/2022]
Abstract
A2B adenosine receptor (A2BAR) antagonists have therapeutic potential in inflammation-related diseases such as asthma, chronic obstructive pulmonary disease and cancer. However, no drug is currently clinically approved, creating a demand for research on novel antagonists. Over the last decade, the study of target binding kinetics, along with affinity and potency, has been proven valuable in early drug discovery stages, as it is associated with improved in vivo drug efficacy and safety. In this study, we report the synthesis and biological evaluation of a series of xanthine derivatives as A2BAR antagonists, including an isothiocyanate derivative designed to bind covalently to the receptor. All 28 final compounds were assessed in radioligand binding experiments, to evaluate their affinity and for those qualifying, kinetic binding parameters. Both structure-affinity and structure-kinetic relationships were derived, providing a clear relationship between affinity and dissociation rate constants. Two structurally similar compounds, 17 and 18, were further evaluated in a label-free assay due to their divergent kinetic profiles. An extended cellular response was associated with long A2BAR residence times. This link between a ligand's A2BAR residence time and its functional effect highlights the importance of binding kinetics as a selection parameter in the early stages of drug discovery.
Collapse
Affiliation(s)
- Anna Vlachodimou
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, the Netherlands
| | - Henk de Vries
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, the Netherlands
| | - Milena Pasoli
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, the Netherlands
| | - Miranda Goudswaard
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, the Netherlands
| | - Soon-Ai Kim
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Yong-Chul Kim
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Mirko Scortichini
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Melissa Marshall
- Department of Internal Medicine and Molecular Physiology & Biological Physics, University of Virginia Health Science Center, Charlottesville, VA 22908, USA
| | - Joel Linden
- Department of Internal Medicine and Molecular Physiology & Biological Physics, University of Virginia Health Science Center, Charlottesville, VA 22908, USA
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, the Netherlands; Oncode Institute, Leiden, the Netherlands
| | - Kenneth A Jacobson
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| | - Adriaan P IJzerman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, the Netherlands.
| |
Collapse
|
8
|
Thom C, Ehrenmann J, Vacca S, Waltenspühl Y, Schöppe J, Medalia O, Plückthun A. Structures of neurokinin 1 receptor in complex with G q and G s proteins reveal substance P binding mode and unique activation features. SCIENCE ADVANCES 2021; 7:eabk2872. [PMID: 34878828 PMCID: PMC8654284 DOI: 10.1126/sciadv.abk2872] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 10/18/2021] [Indexed: 06/13/2023]
Abstract
The neurokinin 1 receptor (NK1R) is involved in inflammation and pain transmission. This pathophysiologically important G protein–coupled receptor is predominantly activated by its cognate agonist substance P (SP) but also by the closely related neurokinins A and B. Here, we report cryo–electron microscopy structures of SP-bound NK1R in complex with its primary downstream signal mediators, Gq and Gs. Our structures reveal how a polar network at the extracellular, solvent-exposed receptor surface shapes the orthosteric pocket and that NK1R adopts a noncanonical active-state conformation with an interface for G protein binding, which is distinct from previously reported structures. Detailed comparisons with antagonist-bound NK1R crystal structures reveal that insurmountable antagonists induce a distinct and long-lasting receptor conformation that sterically blocks SP binding. Together, our structures provide important structural insights into ligand and G protein promiscuity, the lack of basal signaling, and agonist- and antagonist-induced conformations in the neurokinin receptor family.
Collapse
|
9
|
Arsova A, Møller TC, Hellyer SD, Vedel L, Foster SR, Hansen JL, Bräuner-Osborne H, Gregory KJ. Positive Allosteric Modulators of Metabotropic Glutamate Receptor 5 as Tool Compounds to Study Signaling Bias. Mol Pharmacol 2021; 99:328-341. [PMID: 33602724 DOI: 10.1124/molpharm.120.000185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/27/2021] [Indexed: 11/22/2022] Open
Abstract
Positive allosteric modulation of metabotropic glutamate subtype 5 (mGlu5) receptor has emerged as a potential new therapeutic strategy for the treatment of schizophrenia and cognitive impairments. However, positive allosteric modulator (PAM) agonist activity has been associated with adverse side effects, and neurotoxicity has also been observed for pure PAMs. The structural and pharmacological basis of therapeutic versus adverse mGlu5 PAM in vivo effects remains unknown. Thus, gaining insights into the signaling fingerprints, as well as the binding kinetics of structurally diverse mGlu5 PAMs, may help in the rational design of compounds with desired properties. We assessed the binding and signaling profiles of N-methyl-5-(phenylethynyl)pyrimidin-2-amine (MPPA), 3-cyano-N-(2,5-diphenylpyrazol-3-yl)benzamide (CDPPB), and 1-[4-(4-chloro-2-fluoro-phenyl)piperazin-1-yl]-2-(4-pyridylmethoxy)ethenone [compound 2c, a close analog of 1-(4-(2-chloro-4-fluorophenyl)piperazin-1-yl)-2-(pyridin-4-ylmethoxy)ethanone] in human embryonic kidney 293A cells stably expressing mGlu5 using Ca2+ mobilization, inositol monophosphate (IP1) accumulation, extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation, and receptor internalization assays. Of the three allosteric ligands, only CDPPB had intrinsic agonist efficacy, and it also had the longest receptor residence time and highest affinity. MPPA was a biased PAM, showing higher positive cooperativity with orthosteric agonists in ERK1/2 phosphorylation and Ca2+ mobilization over IP1 accumulation and receptor internalization. In primary cortical neurons, all three PAMs showed stronger positive cooperativity with (S)-3,5-dihydroxyphenylglycine (DHPG) in Ca2+ mobilization over IP1 accumulation. Our characterization of three structurally diverse mGlu5 PAMs provides further molecular pharmacological insights and presents the first assessment of PAM-mediated mGlu5 internalization. SIGNIFICANCE STATEMENT: Enhancing metabotropic glutamate receptor subtype 5 (mGlu5) activity is a promising strategy to treat cognitive and positive symptoms in schizophrenia. It is increasingly evident that positive allosteric modulators (PAMs) of mGlu5 are not all equal in preclinical models; there remains a need to better understand the molecular pharmacological properties of mGlu5 PAMs. This study reports detailed characterization of the binding and functional pharmacological properties of mGlu5 PAMs and is the first study of the effects of mGlu5 PAMs on receptor internalization.
Collapse
Affiliation(s)
- Angela Arsova
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, Australia (S.D.H., K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Novo Nordisk Park 1, Måløv, Denmark (J.L.H.)
| | - Thor C Møller
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, Australia (S.D.H., K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Novo Nordisk Park 1, Måløv, Denmark (J.L.H.)
| | - Shane D Hellyer
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, Australia (S.D.H., K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Novo Nordisk Park 1, Måløv, Denmark (J.L.H.)
| | - Line Vedel
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, Australia (S.D.H., K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Novo Nordisk Park 1, Måløv, Denmark (J.L.H.)
| | - Simon R Foster
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, Australia (S.D.H., K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Novo Nordisk Park 1, Måløv, Denmark (J.L.H.)
| | - Jakob L Hansen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, Australia (S.D.H., K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Novo Nordisk Park 1, Måløv, Denmark (J.L.H.)
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, Australia (S.D.H., K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Novo Nordisk Park 1, Måløv, Denmark (J.L.H.)
| | - Karen J Gregory
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, Australia (S.D.H., K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Novo Nordisk Park 1, Måløv, Denmark (J.L.H.)
| |
Collapse
|
10
|
Bertolini M, McElwee K, Gilhar A, Bulfone‐Paus S, Paus R. Hair follicle immune privilege and its collapse in alopecia areata. Exp Dermatol 2020; 29:703-725. [DOI: 10.1111/exd.14155] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/18/2020] [Accepted: 07/10/2020] [Indexed: 12/11/2022]
Affiliation(s)
| | - Kevin McElwee
- Monasterium Laboratory Münster Germany
- Centre for Skin Sciences University of Bradford Bradford UK
- Department of Dermatology and Skin Science University of British Columbia Vancouver British Columbia Canada
| | - Amos Gilhar
- Laboratory for Skin Research Rappaport Faculty of Medicine Technion‐Israel Institute of Technology Haifa Israel
| | - Silvia Bulfone‐Paus
- Monasterium Laboratory Münster Germany
- Centre for Dermatology Research University of Manchester and NIHR Manchester Biomedical Research Centre Manchester UK
| | - Ralf Paus
- Monasterium Laboratory Münster Germany
- Centre for Dermatology Research University of Manchester and NIHR Manchester Biomedical Research Centre Manchester UK
- Dr. Philip Frost Department of Dermatology & Cutaneous Surgery University of Miami Miller School of Medicine Miami FL USA
| |
Collapse
|
11
|
Arsova A, Møller TC, Vedel L, Hansen JL, Foster SR, Gregory KJ, Bräuner-Osborne H. Detailed In Vitro Pharmacological Characterization of Clinically Tested Negative Allosteric Modulators of the Metabotropic Glutamate Receptor 5. Mol Pharmacol 2020; 98:49-60. [PMID: 32358164 PMCID: PMC7705108 DOI: 10.1124/mol.119.119032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/10/2020] [Indexed: 12/14/2022] Open
Abstract
Negative allosteric modulation of the metabotropic glutamate 5 (mGlu5) receptor has emerged as a potential strategy for the treatment of neurologic disorders. Despite the success in preclinical studies, many mGlu5 negative allosteric modulators (NAMs) that have reached clinical trials failed due to lack of efficacy. In this study, we provide a detailed in vitro pharmacological characterization of nine clinically and preclinically tested NAMs. We evaluated inhibition of l-glutamate-induced signaling with Ca2+ mobilization, inositol monophosphate (IP1) accumulation, extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation, and real-time receptor internalization assays on rat mGlu5 expressed in HEK293A cells. Moreover, we determined association rates (kon) and dissociation rates (koff), as well as NAM affinities with [3H]methoxy-PEPy binding experiments. kon and koff values varied greatly between the nine NAMs (34- and 139-fold, respectively) resulting in long receptor residence times (>400 min) for basimglurant and mavoglurant, medium residence times (10-30 min) for AZD2066, remeglurant, and (RS)-remeglurant, and low residence times (<10 mins) for dipraglurant, F169521, F1699611, and STX107. We found that all NAMs inhibited l-glutamate-induced mGlu5 receptor internalization, generally with a similar potency to IP1 accumulation and ERK1/2 phosphorylation, whereas Ca2+ mobilization was less potently inhibited. Operational model of allosterism analyses revealed that dipraglurant and (RS)-remeglurant were biased toward (affinity) receptor internalization and away (cooperativity) from the ERK1/2 phosphorylation pathway, respectively. Our study is the first to measure mGlu5 NAM binding kinetics and negative allosteric modulation of mGlu5 receptor internalization and adds significant new knowledge about the molecular pharmacology of a diverse range of clinically relevant NAMs. SIGNIFICANCE STATEMENT: The metabotropic glutamate 5 (mGlu5) receptor is important in many brain functions and implicated in several neurological pathologies. Negative allosteric modulators (NAMs) have shown promising results in preclinical models but have so far failed in human clinical trials. Here we provide the most comprehensive and comparative molecular pharmacological study to date of nine preclinically/clinically tested NAMs at the mGlu5 receptor, which is also the first study to measure ligand binding kinetics and negative allosteric modulation of mGlu5 receptor internalization.
Collapse
Affiliation(s)
- Angela Arsova
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Måløv, Denmark (J.L.H.)
| | - Thor C Møller
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Måløv, Denmark (J.L.H.)
| | - Line Vedel
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Måløv, Denmark (J.L.H.)
| | - Jakob Lerche Hansen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Måløv, Denmark (J.L.H.)
| | - Simon R Foster
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Måløv, Denmark (J.L.H.)
| | - Karen J Gregory
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Måløv, Denmark (J.L.H.)
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Måløv, Denmark (J.L.H.)
| |
Collapse
|
12
|
van der Velden WJC, Heitman LH, Rosenkilde MM. Perspective: Implications of Ligand-Receptor Binding Kinetics for Therapeutic Targeting of G Protein-Coupled Receptors. ACS Pharmacol Transl Sci 2020; 3:179-189. [PMID: 32296761 DOI: 10.1021/acsptsci.0c00012] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Indexed: 12/16/2022]
Abstract
The concept of ligand-receptor binding kinetics has been broadly applied in drug development pipelines focusing on G protein-coupled receptors (GPCRs). The ligand residence time (RT) for a receptor describes how long a ligand-receptor complex exists, and is defined as the reciprocal of the dissociation rate constant (k off). RT has turned out to be a valuable parameter for GPCR researchers focusing on drug development as a good predictor of in vivo efficacy. The positive correlation between RT and in vivo efficacy has been established for several drugs targeting class A GPCRs (e.g., the neurokinin-1 receptor (NK1R), the β2 adrenergic receptor (β2AR), and the muscarinic 3 receptor (M3R)) and for drugs targeting class B1 (e.g., the glucagon-like peptide 1 receptor (GLP-1R)). Recently, the association rate constant (k on) has gained similar attention as another parameter affecting in vivo efficacy. In the current perspective, we address the importance of studying ligand-receptor binding kinetics for therapeutic targeting of GPCRs, with an emphasis on how binding kinetics can be altered by subtle molecular changes in the ligands and/or the receptors and how such changes affect treatment outcome. Moreover, we speculate on the impact of binding kinetic parameters for functional selectivity and sustained receptor signaling from endosomal compartments; phenomena that have gained increasing interest in attempts to improve therapeutic targeting of GPCRs.
Collapse
Affiliation(s)
- Wijnand J C van der Velden
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK 2200, Denmark
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2333 CC, The Netherlands
| | - Mette M Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK 2200, Denmark
| |
Collapse
|
13
|
Vincenzi B, Trower M, Duggal A, Guglielmini P, Harris P, Jackson D, Lacouture ME, Ratti E, Tonini G, Wood A, Ständer S. Neurokinin-1 antagonist orvepitant for EGFRI-induced pruritus in patients with cancer: a randomised, placebo-controlled phase II trial. BMJ Open 2020; 10:e030114. [PMID: 32034016 PMCID: PMC7045265 DOI: 10.1136/bmjopen-2019-030114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 12/11/2019] [Accepted: 01/13/2020] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE To evaluate the efficacy of orvepitant (10 or 30 mg given once daily, orally for 4 weeks), a neurokinin-1 receptor antagonist, compared with placebo in reducing the intensity of epidermal growth factor receptor inhibitor (EGFRI)-induced intense pruritus. DESIGN Randomised, double-blind, placebo-controlled clinical trial. SETTING 15 hospitals in Italy and five hospitals in the UK. PARTICIPANTS 44 patients aged ≥18 years receiving an EGFRI for a histologically confirmed malignant solid tumour and experiencing moderate or intense pruritus after EGFRI treatment. INTERVENTION 30 or 10 mg orvepitant or placebo tablets once daily for 4 weeks (randomised 1:1:1). PRIMARY AND SECONDARY OUTCOME MEASURES The primary endpoint was change from baseline in mean patient-recorded numerical rating scale (NRS) score (over the last three recordings) at week 4. Secondary outcome measures were NRS score, verbal rating scale score, Skindex-16 and Leeds Sleep Evaluation Questionnaire at each study visit (baseline, weeks 1, 4, 8); rescue medication use; EGFRI dose reduction; and study withdrawal because of intense uncontrolled pruritus. RESULTS The trial was terminated early because of recruitment challenges; only 44 of the planned 90 patients were randomised. All patients were analysed for efficacy and safety. Mean NRS score change from baseline to week 4 was -2.78 (SD: 2.64) points in the 30 mg group, -3.04 (SD: 3.06) points in the 10 mg group and -3.21 (SD: 1.77) points in the placebo group; the difference between orvepitant and placebo was not statistically significant. No safety signal was detected. Adverse events related to orvepitant (asthenia, dizziness, dry mouth, hyperhidrosis) were all of mild or moderate severity. CONCLUSIONS Orvepitant was safe and well tolerated. No difference in NRS score between the orvepitant and placebo groups was observed at the week 4 primary endpoint. A number of explanations for this outcome are possible. TRIAL REGISTRATION NUMBER EudraCT2013-002763-25.
Collapse
Affiliation(s)
- Bruno Vincenzi
- Medical Oncology, Universita Campus Bio-Medico di Roma Facolta di Medicina e Chirurgia, Roma, Italy
| | | | - Ajay Duggal
- Adnovate Clinical Development Strategies, East Sussex, UK
| | | | | | | | - Mario E Lacouture
- Department of Dermatology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Giuseppe Tonini
- Medical Oncology, Universita Campus Bio-Medico di Roma Facolta di Medicina e Chirurgia, Roma, Italy
| | | | - Sonja Ständer
- Center for Chronic Pruritus, University Hospital Münster, Münster, Germany
| |
Collapse
|
14
|
Laprairie RB, Vemuri K, Stahl EL, Korde A, Ho JH, Grim TW, Hua T, Wu Y, Stevens RC, Liu ZJ, Makriyannis A, Bohn LM. Probing the CB 1 Cannabinoid Receptor Binding Pocket with AM6538, a High-Affinity Irreversible Antagonist. Mol Pharmacol 2019; 96:619-628. [PMID: 31515283 PMCID: PMC6785652 DOI: 10.1124/mol.119.116483] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 08/17/2019] [Indexed: 01/12/2023] Open
Abstract
Cannabinoid receptor 1 (CB1) is a potential therapeutic target for the treatment of pain, obesity and obesity-related metabolic disorders, and addiction. The crystal structure of human CB1 has been determined in complex with the stabilizing antagonist AM6538. In the present study, we characterize AM6538 as a tight-binding/irreversible antagonist of CB1, as well as two derivatives of AM6538 (AM4112 and AM6542) as slowly dissociating CB1 antagonists across binding simulations and cellular signaling assays. The long-lasting nature of AM6538 was explored in vivo wherein AM6538 continues to block CP55,940-mediated behaviors in mice up to 5 days after a single injection. In contrast, the effects of SR141716A abate in mice 2 days after injection. These studies demonstrate the functional outcome of CB1 antagonist modification and open the path for development of long-lasting CB1 antagonists.
Collapse
Affiliation(s)
- Robert B Laprairie
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, Florida (R.B.L., E.L.S., J.-H.H., T.W.G., L.M.B.); Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (K.V., A.K., A.M.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H., Y.W., Z.-J.L.); and Departments of Biological Sciences and Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California (R.C.S.)
| | - Kiran Vemuri
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, Florida (R.B.L., E.L.S., J.-H.H., T.W.G., L.M.B.); Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (K.V., A.K., A.M.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H., Y.W., Z.-J.L.); and Departments of Biological Sciences and Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California (R.C.S.)
| | - Edward L Stahl
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, Florida (R.B.L., E.L.S., J.-H.H., T.W.G., L.M.B.); Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (K.V., A.K., A.M.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H., Y.W., Z.-J.L.); and Departments of Biological Sciences and Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California (R.C.S.)
| | - Anisha Korde
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, Florida (R.B.L., E.L.S., J.-H.H., T.W.G., L.M.B.); Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (K.V., A.K., A.M.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H., Y.W., Z.-J.L.); and Departments of Biological Sciences and Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California (R.C.S.)
| | - Jo-Hao Ho
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, Florida (R.B.L., E.L.S., J.-H.H., T.W.G., L.M.B.); Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (K.V., A.K., A.M.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H., Y.W., Z.-J.L.); and Departments of Biological Sciences and Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California (R.C.S.)
| | - Travis W Grim
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, Florida (R.B.L., E.L.S., J.-H.H., T.W.G., L.M.B.); Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (K.V., A.K., A.M.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H., Y.W., Z.-J.L.); and Departments of Biological Sciences and Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California (R.C.S.)
| | - Tian Hua
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, Florida (R.B.L., E.L.S., J.-H.H., T.W.G., L.M.B.); Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (K.V., A.K., A.M.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H., Y.W., Z.-J.L.); and Departments of Biological Sciences and Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California (R.C.S.)
| | - Yiran Wu
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, Florida (R.B.L., E.L.S., J.-H.H., T.W.G., L.M.B.); Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (K.V., A.K., A.M.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H., Y.W., Z.-J.L.); and Departments of Biological Sciences and Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California (R.C.S.)
| | - Raymond C Stevens
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, Florida (R.B.L., E.L.S., J.-H.H., T.W.G., L.M.B.); Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (K.V., A.K., A.M.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H., Y.W., Z.-J.L.); and Departments of Biological Sciences and Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California (R.C.S.)
| | - Zhi-Jie Liu
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, Florida (R.B.L., E.L.S., J.-H.H., T.W.G., L.M.B.); Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (K.V., A.K., A.M.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H., Y.W., Z.-J.L.); and Departments of Biological Sciences and Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California (R.C.S.)
| | - Alexandros Makriyannis
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, Florida (R.B.L., E.L.S., J.-H.H., T.W.G., L.M.B.); Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (K.V., A.K., A.M.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H., Y.W., Z.-J.L.); and Departments of Biological Sciences and Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California (R.C.S.)
| | - Laura M Bohn
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, Jupiter, Florida (R.B.L., E.L.S., J.-H.H., T.W.G., L.M.B.); Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (K.V., A.K., A.M.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H., Y.W., Z.-J.L.); and Departments of Biological Sciences and Chemistry, Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California (R.C.S.)
| |
Collapse
|
15
|
Gilhar A, Laufer-Britva R, Keren A, Paus R. Frontiers in alopecia areata pathobiology research. J Allergy Clin Immunol 2019; 144:1478-1489. [PMID: 31606262 DOI: 10.1016/j.jaci.2019.08.035] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 12/11/2022]
Abstract
This current review explores selected and as yet insufficiently investigated frontiers in current alopecia areata (AA) pathobiology research, with an emphasis on potential "new" players in AA pathobiology that deserve more systematic exploration and therapeutic targeting. Indeed, new evidence suggests that CD8+ T cells, which have long been thought to be the central players in AA pathobiology, are not the only drivers of disease. Instead, subsets of natural killer (NK) and so-called "unconventional" T cells (invariant NK T cells, γδ T cells, classic NK cells, and type 1 innate lymphoid cells), all of which can produce large amounts of IFN-γ, might also drive AA pathobiology independent of classical, autoantigen-dependent CD8+ T-cell functions. Another important new frontier is the role of regulatory lymphocyte subsets, such as regulatory T cells, γδ regulatory T cells, NKT10 cells, and perifollicular mast cells, in maintaining physiologic hair follicle immune privilege (IP); the extent to which these functions are defective in patients with AA; and how this IP-protective role could be restored therapeutically in patients with established AA. Broadening our AA research horizon along the lines suggested above promises not only to open the door to innovative and even more effective immunotherapy strategies for AA but will also likely be relevant for other autoimmune disorders in which pathobiology, ectopic MHC class I expression, and IP collapse play an important role.
Collapse
Affiliation(s)
- Amos Gilhar
- Skin Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| | | | - Aviad Keren
- Skin Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ralf Paus
- Dr Philipp Frost Department of Dermatology & Cutaneous Surgery, Miller School of Medicine, University of Miami, Miami, Fla; Dermatology Research Centre, University of Manchester and NIHR Manchester Biomedical Research Centre, Manchester, United Kingdom
| |
Collapse
|
16
|
Mocking TAM, Buzink MCML, Leurs R, Vischer HF. Bioluminescence Resonance Energy Transfer Based G Protein-Activation Assay to Probe Duration of Antagonism at the Histamine H 3 Receptor. Int J Mol Sci 2019; 20:ijms20153724. [PMID: 31366084 PMCID: PMC6695674 DOI: 10.3390/ijms20153724] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/27/2019] [Accepted: 07/28/2019] [Indexed: 12/30/2022] Open
Abstract
Duration of receptor antagonism, measured as the recovery of agonist responsiveness, is gaining attention as a method to evaluate the 'effective' target-residence for antagonists. These functional assays might be a good alternative for kinetic binding assays in competition with radiolabeled or fluorescent ligands, as they are performed on intact cells and better reflect consequences of dynamic cellular processes on duration of receptor antagonism. Here, we used a bioluminescence resonance energy transfer (BRET)-based assay that monitors heterotrimeric G protein activation via scavenging of released Venus-Gβ1γ2 by NanoLuc (Nluc)-tagged membrane-associated-C-terminal fragment of G protein-coupled receptor kinase 3 (masGRK3ct-Nluc) as a tool to probe duration of G protein-coupled receptor (GPCR) antagonism. The Gαi-coupled histamine H3 receptor (H3R) was used in this study as prolonged antagonism is associated with adverse events (e.g., insomnia) and consequently, short-residence time ligands might be preferred. Due to its fast and prolonged response, this assay can be used to determine the duration of functional antagonism by measuring the recovery of agonist responsiveness upon washout of pre-bound antagonist, and to assess antagonist re-equilibration time via Schild-plot analysis. Re-equilibration of pre-incubated antagonist with agonist and receptor could be followed in time to monitor the transition from insurmountable to surmountable antagonism. The BRET-based G protein activation assay can detect differences in the recovery of H3R responsiveness and re-equilibration of pre-bound antagonists between the tested H3R antagonists. Fast dissociation kinetics were observed for marketed drug pitolisant (Wakix®) in this assay, which suggests that short residence time might be beneficial for therapeutic targeting of the H3R.
Collapse
Affiliation(s)
- Tamara A M Mocking
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Maurice C M L Buzink
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Rob Leurs
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Henry F Vischer
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands.
| |
Collapse
|
17
|
Schöppe J, Ehrenmann J, Klenk C, Rucktooa P, Schütz M, Doré AS, Plückthun A. Crystal structures of the human neurokinin 1 receptor in complex with clinically used antagonists. Nat Commun 2019; 10:17. [PMID: 30604743 PMCID: PMC6318301 DOI: 10.1038/s41467-018-07939-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/03/2018] [Indexed: 12/28/2022] Open
Abstract
Neurokinins (or tachykinins) are peptides that modulate a wide variety of human physiology through the neurokinin G protein-coupled receptor family, implicated in a diverse array of pathological processes. Here we report high-resolution crystal structures of the human NK1 receptor (NK1R) bound to two small-molecule antagonist therapeutics – aprepitant and netupitant and the progenitor antagonist CP-99,994. The structures reveal the detailed interactions between clinically approved antagonists and NK1R, which induce a distinct receptor conformation resulting in an interhelical hydrogen-bond network that cross-links the extracellular ends of helices V and VI. Furthermore, the high-resolution details of NK1R bound to netupitant establish a structural rationale for the lack of basal activity in NK1R. Taken together, these co-structures provide a comprehensive structural basis of NK1R antagonism and will facilitate the design of new therapeutics targeting the neurokinin receptor family. Neurokinin receptors are G protein-coupled receptors. Here the authors present three crystal structures of the neurokinin 1 receptor (NK1R) in complex with small-molecule antagonists including aprepitant and netupitant and observe that these clinically approved compounds induce a conformational change in the receptor.
Collapse
Affiliation(s)
- Jendrik Schöppe
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland
| | - Janosch Ehrenmann
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland
| | - Christoph Klenk
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland
| | - Prakash Rucktooa
- Sosei Heptares, Steinmetz Building, Granta Park, Great Abington, Cambridge, CB21 6DG, UK
| | - Marco Schütz
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland.,Heptares Therapeutics Zürich AG, Grabenstrasse 11a, 8952, Zürich, Switzerland
| | - Andrew S Doré
- Sosei Heptares, Steinmetz Building, Granta Park, Great Abington, Cambridge, CB21 6DG, UK
| | - Andreas Plückthun
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland.
| |
Collapse
|
18
|
The implications of target saturation for the use of drug–target residence time. Nat Rev Drug Discov 2018; 18:82-84. [DOI: 10.1038/nrd.2018.234] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
19
|
Aminoethane Sulfonic Acid Magnesium Salt Inhibits Ca 2+ Entry Through NMDA Receptor Ion Channel In Vitro. Bull Exp Biol Med 2018; 166:39-42. [PMID: 30417298 DOI: 10.1007/s10517-018-4284-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Indexed: 10/27/2022]
Abstract
The effect of a cerebroprotective agent magnesium bis-aminoethanesulfonate (laboratory code FS-LKhT-317) on intracellular calcium concentration was studied by the fluorescent imaging technique on neuroglial cell culture from Spraque-Dawley rat hippocampus. The substance produced a pronounced inhibitory effect and suppressed NMDA receptor activity in concentrations of ≥50 μM. The observed effects were reversible or partially reversible and were detected by a decrease in Ca2+ signal amplitude in neurons in response to NMDA applications in a Mg2+-free medium and by inhibition of Ca2+ pulses in magnesium-free medium (elimination of magnesium block).
Collapse
|
20
|
Henssen AG, Odersky A, Szymansky A, Seiler M, Althoff K, Beckers A, Speleman F, Schäfers S, De Preter K, Astrahanseff K, Struck J, Schramm A, Eggert A, Bergmann A, Schulte JH. Targeting tachykinin receptors in neuroblastoma. Oncotarget 2018; 8:430-443. [PMID: 27888795 PMCID: PMC5352132 DOI: 10.18632/oncotarget.13440] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 11/12/2016] [Indexed: 01/23/2023] Open
Abstract
Neuroblastoma is the most common extracranial tumor in children. Despite aggressive multimodal treatment, high-risk neuroblastoma remains a clinical challenge with survival rates below 50%. Adding targeted drugs to first-line therapy regimens is a promising approach to improve survival in these patients. TACR1 activation by substance P has been reported to be mitogenic in cancer cell lines. Tachykinin receptor (TACR1) antagonists are approved for clinical use as an antiemetic remedy since 2003. Tachykinin receptor inhibition has recently been shown to effectively reduce growth of several tumor types. Here, we report that neuroblastoma cell lines express TACR1, and that targeting TACR1 activity significantly reduced cell viability and induced apoptosis in neuroblastoma cell lines. Gene expression profiling revealed that TACR1 inhibition repressed E2F2 and induced TP53 signaling. Treating mice harboring established neuroblastoma xenograft tumors with Aprepitant also significantly reduced tumor burden. Thus, we provide evidence that the targeted inhibition of tachykinin receptor signaling shows therapeutic efficacy in preclinical models for high-risk neuroblastoma.
Collapse
Affiliation(s)
- Anton G Henssen
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Andrea Odersky
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Germany
| | - Annabell Szymansky
- Department of Pediatric Oncology/Hematology, Charité- Universitätsmedizin Berlin, Germany
| | | | - Kristina Althoff
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Germany
| | - Anneleen Beckers
- Center of Medical Genetics Ghent (CMGG), Ghent University Hospital, Belgium
| | - Frank Speleman
- Center of Medical Genetics Ghent (CMGG), Ghent University Hospital, Belgium
| | - Simon Schäfers
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Germany
| | - Katleen De Preter
- Center of Medical Genetics Ghent (CMGG), Ghent University Hospital, Belgium
| | - Kathy Astrahanseff
- Department of Pediatric Oncology/Hematology, Charité- Universitätsmedizin Berlin, Germany
| | | | - Alexander Schramm
- Department of Pediatric Oncology and Hematology, University Children's Hospital Essen, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology/Hematology, Charité- Universitätsmedizin Berlin, Germany.,German Consortium for Translational Cancer Research (DKTK), Partner Site Charite Berlin, Berlin, Germany
| | | | - Johannes H Schulte
- Department of Pediatric Oncology/Hematology, Charité- Universitätsmedizin Berlin, Germany.,German Consortium for Translational Cancer Research (DKTK), Partner Site Charite Berlin, Berlin, Germany
| |
Collapse
|
21
|
Morrey ME, Sanchez-Sotelo J, Lewallen EA, An KN, Grill DE, Steinmann SP, Yao JJ, Salib CG, Trousdale WH, Reina N, Kremers HM, Lewallen DG, van Wijnen AJ, Abdel MP. Intra-articular injection of a substance P inhibitor affects gene expression in a joint contracture model. J Cell Biochem 2018; 119:1326-1336. [PMID: 28671282 PMCID: PMC6388635 DOI: 10.1002/jcb.26256] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 06/30/2017] [Indexed: 01/23/2023]
Abstract
Substance P (SP), a neurotransmitter released after injury, has been linked to deregulated tissue repair and fibrosis in musculoskeletal tissues and other organs. Although SP inhibition is an effective treatment for nausea, it has not been previously considered as an anti-fibrotic therapy. Although there are extensive medical records of individuals who have used SP antagonists, our analysis of human registry data revealed that patients receiving these antagonists and arthroplasty are exceedingly rare, thus precluding a clinical evaluation of their potential effects in the context of arthrofibrosis. Therefore, we pursued in vivo studies to assess the effect of SP inhibition early after injury on pro-fibrotic gene expression and contractures in an animal model of post-traumatic joint stiffening. Skeletally mature rabbits (n = 24) underwent surgically induced severe joint contracture, while injected with either fosaprepitant (a selective SP antagonist) or saline (control) early after surgery (3, 6, 12, and 24 h). Biomechanical testing revealed that differences in mean contracture angles between the groups were not statistically significant (P = 0.27), suggesting that the drug neither mitigates nor exacerbates joint contracture. However, microarray gene expression analysis revealed that mRNA levels for proteins related to cell signaling, pro-angiogenic, pro-inflammatory, and collagen matrix production were significantly different between control and fosaprepitant treated rabbits (P < 0.05). Hence, our study demonstrates that inhibition of SP alters expression of pro-fibrotic genes in vivo. This finding will motivate future studies to optimize interventions that target SP to reduce the formation of post-traumatic joint contractures.
Collapse
Affiliation(s)
- Mark E. Morrey
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
| | | | | | - Kai-Nan An
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
| | - Diane E. Grill
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | | | - Jie J. Yao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
| | | | | | - Nicolas Reina
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
| | - Hilal M. Kremers
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
- Health Sciences Research, Mayo Clinic, Rochester, MN
| | | | | | | |
Collapse
|
22
|
Paus R, Bulfone-Paus S, Bertolini M. Hair Follicle Immune Privilege Revisited: The Key to Alopecia Areata Management. J Investig Dermatol Symp Proc 2018; 19:S12-S17. [PMID: 29273098 DOI: 10.1016/j.jisp.2017.10.014] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The collapse of the immune privilege (IP) of the anagen hair bulb is now accepted as a key element in AA pathogenesis, and hair bulb IP restoration lies at the core of AA therapy. Here, we briefly review the essentials of hair bulb IP and recent progress in understanding its complexity. We discuss open questions and why the systematic dissection of hair bulb IP and its pharmacological manipulation (including the clinical testing of FK506 and α-melanocyte-stimulating hormone analogs) promise to extend the range of future therapeutic options in AA and other IP collapse-related autoimmune diseases.
Collapse
Affiliation(s)
- Ralf Paus
- Centre for Dermatology Research, University of Manchester, NIHR Manchester Biomedical Research Centre and MAHSC, Manchester, UK.
| | - Silvia Bulfone-Paus
- Centre for Dermatology Research, University of Manchester, NIHR Manchester Biomedical Research Centre and MAHSC, Manchester, UK
| | - Marta Bertolini
- Department of Dermatology, University of Münster, Münster, Germany
| |
Collapse
|
23
|
Choi H, Kim DJ, Nam S, Lim S, Hwang JS, Park KS, Hong HS, Won Y, Shin MK, Chung E, Son Y. Substance P restores normal skin architecture and reduces epidermal infiltration of sensory nerve fiber in TNCB-induced atopic dermatitis-like lesions in NC/Nga mice. J Dermatol Sci 2017; 89:248-257. [PMID: 29269174 DOI: 10.1016/j.jdermsci.2017.11.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 11/24/2017] [Accepted: 11/28/2017] [Indexed: 11/19/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic inflammatory skin disorder characterized by intense pruritus and eczematous lesion. Substance P (SP) is an 11-amino-acid endogenous neuropeptide that belongs to the tachykinin family and several reports recently have supported the anti-inflammatory and tissue repairing roles of SP. OBJECTIVE In this study, we investigated whether SP can improve AD symptoms, especially the impaired skin barrier function, in 2, 4, 6-trinitrochlorobenzene (TNCB)-induced chronic dermatitis of NC/Nga mice or not. METHOD AD-like dermatitis was induced in NC/Nga mice by repeated sensitization with TNCB for 5 weeks. The experimental group designations and topical treatments were as follows: vehicle group (AD-VE); SP group (AD-SP); and SP with NK1R antagonist CP99994 (AD-SP-A) group. Histological analysis was performed to evaluate epidermal differentiation, dermal integrity, and epidermal nerve innervation in AD-like lesions. The skin barrier functions and pruritus of NC/Nga mice were evaluated by measuring transepidermal water loss (TEWL) and scratching behavior, respectively. RESULT Topical SP treatment resulted in significant down-regulation of Ki67 and the abnormal-type keratins (K) K6, K16, and K17, restoration of filaggrin and claudin-1, marked reduction of TEWL, and restoration of basement membrane and dermal collagen deposition, even under continuous sensitization of low dose TNCB. In addition, SP significantly reduced innervation of itch-evoking nerve fibers, gelatinase activity and nerve growth factor (NGF) expression in the epidermis but upregulated semaphorin-3A (Sema3A) expression in the epidermis, along with reduced scratching behavior in TNCB-treated NC/Nga mice. All of these effects were completely reversed by co-treatment with the NK1R antagonist CP99994. In cultured human keratinocytes, SP treatment reduced expression of TGF-α, but upregulated TGF-β and Sema3A. CONCLUSION Topically administered SP can restore normal skin barrier function, reduce epidermal infiltration of itch-evoking nerve fibers in the AD-like skin lesions, and alleviate scratching behavior. Thus, SP may be proposed as a potential medication for chronic dermatitis and AD.
Collapse
Affiliation(s)
- Hyeongwon Choi
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Yong In 446-701, Republic of Korea
| | - Dong-Jin Kim
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Yong In 446-701, Republic of Korea
| | - Seungwoo Nam
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Yong In 446-701, Republic of Korea
| | - Sunki Lim
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Yong In 446-701, Republic of Korea
| | - Jae-Sung Hwang
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Yong In 446-701, Republic of Korea
| | - Ki Sook Park
- East-West Medical Research Institute, College of Medicine, Kyung Hee University, Republic of Korea; Kyung Hee Institute of Regenerative Medicine, Republic of Korea
| | - Hyun Sook Hong
- East-West Medical Research Institute, College of Medicine, Kyung Hee University, Republic of Korea; Kyung Hee Institute of Regenerative Medicine, Republic of Korea
| | - Younsun Won
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Yong In 446-701, Republic of Korea
| | - Min Kyung Shin
- Department of Dermatology, Kyung Hee University Hospital, Seoul, Republic of Korea.
| | - Eunkyung Chung
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Yong In 446-701, Republic of Korea; BIO R&D Center, L&K BIOMED CO. LTD., Seoul, Republic of Korea.
| | - Youngsook Son
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Yong In 446-701, Republic of Korea; Kyung Hee Institute of Regenerative Medicine, Republic of Korea.
| |
Collapse
|
24
|
Choi H, Kim DJ, Nam S, Lim S, Hwang JS, Park KS, Hong HS, Shin MK, Chung E, Son Y. Manifestation of atopic dermatitis-like skin in TNCB-induced NC/Nga mice is ameliorated by topical treatment of substance P, possibly through blockade of allergic inflammation. Exp Dermatol 2017; 27:396-402. [PMID: 28833499 DOI: 10.1111/exd.13421] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2017] [Indexed: 01/11/2023]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disorder characterized by intense pruritus and eczematous lesion. In this study, topically applied substance P (SP) significantly alleviated AD-like clinical symptoms in 2, 4, 6-trinitrochlorobenzene (TNCB)-induced dermatitis in NC/Nga mice. This effect was nullified by pretreatment of the neurokinin-1 receptor (NK-1R) antagonist CP99994. SP treatment significantly reduced the infiltration of mast cells and CD3-positive T cells as well as inflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and thymic stromal lymphopoietin (TSLP), in AD-like skin lesions and decreased the levels of IgE and thymus and activation-regulated chemokine in serum. This SP-induced alleviation of allergic inflammatory responses was also confirmed as reduced activation in the axillary lymph nodes (aLN) and spleen, suggesting the systemic effect of SP on immune responses in TNCB-induced NC/Nga mice. Furthermore, SP-mediated TSLP reduction was confirmed in human keratinocyte culture under pro-inflammatory TNF-α stimulation. Taken together, these results suggest that topically administered SP may have potential as a medication for atopic dermatitis.
Collapse
Affiliation(s)
- Hyeongwon Choi
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Yong In, Korea.,R&D center, Cell & Bio, Seoul, Korea
| | - Dong-Jin Kim
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Yong In, Korea
| | - Seungwoo Nam
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Yong In, Korea.,R&D center, Cell & Bio, Seoul, Korea
| | - Sunki Lim
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Yong In, Korea.,R&D center, Cell & Bio, Seoul, Korea
| | - Jae-Sung Hwang
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Yong In, Korea
| | - Ki Sook Park
- Department of Clinical Pharmacology, College of Medicine, Kyung Hee University, Yong In, Korea.,Kyung Hee Institute of Regenerative Medicine, Dongdaemun-gu, Korea
| | - Hyun Sook Hong
- Department of Clinical Pharmacology, College of Medicine, Kyung Hee University, Yong In, Korea.,Kyung Hee Institute of Regenerative Medicine, Dongdaemun-gu, Korea
| | - Min Kyung Shin
- Department of Dermatology, Kyung Hee University Hospital, Seoul, Korea
| | - Eunkyung Chung
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Yong In, Korea.,Bio R&D center, L&K Biomed Co. Ltd., Seoul, Korea
| | - Youngsook Son
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Yong In, Korea.,Kyung Hee Institute of Regenerative Medicine, Dongdaemun-gu, Korea
| |
Collapse
|
25
|
Nederpelt I, Kuzikov M, de Witte WEA, Schnider P, Tuijt B, Gul S, IJzerman AP, de Lange ECM, Heitman LH. From receptor binding kinetics to signal transduction; a missing link in predicting in vivo drug-action. Sci Rep 2017; 7:14169. [PMID: 29075004 PMCID: PMC5658448 DOI: 10.1038/s41598-017-14257-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 10/09/2017] [Indexed: 11/12/2022] Open
Abstract
An important question in drug discovery is how to overcome the significant challenge of high drug attrition rates due to lack of efficacy and safety. A missing link in the understanding of determinants for drug efficacy is the relation between drug-target binding kinetics and signal transduction, particularly in the physiological context of (multiple) endogenous ligands. We hypothesized that the kinetic binding parameters of both drug and endogenous ligand play a crucial role in determining cellular responses, using the NK1 receptor as a model system. We demonstrated that the binding kinetics of both antagonists (DFA and aprepitant) and endogenous agonists (NKA and SP) have significantly different effects on signal transduction profiles, i.e. potency values, in vitro efficacy values and onset rate of signal transduction. The antagonistic effects were most efficacious with slowly dissociating aprepitant and slowly associating NKA while the combination of rapidly dissociating DFA and rapidly associating SP had less significant effects on the signal transduction profiles. These results were consistent throughout different kinetic assays and cellular backgrounds. We conclude that knowledge of the relationship between in vitro drug-target binding kinetics and cellular responses is important to ultimately improve the understanding of drug efficacy in vivo.
Collapse
Affiliation(s)
- Indira Nederpelt
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Maria Kuzikov
- Fraunhofer IME Screening Port, Schnackenburgallee 114, D-22525, Hamburg, Germany
| | - Wilbert E A de Witte
- Division of Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Patrick Schnider
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Bruno Tuijt
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Sheraz Gul
- Fraunhofer IME Screening Port, Schnackenburgallee 114, D-22525, Hamburg, Germany
| | - Adriaan P IJzerman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Elizabeth C M de Lange
- Division of Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Laura H Heitman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands.
| |
Collapse
|
26
|
Bosma R, Witt G, Vaas LAI, Josimovic I, Gribbon P, Vischer HF, Gul S, Leurs R. The Target Residence Time of Antihistamines Determines Their Antagonism of the G Protein-Coupled Histamine H1 Receptor. Front Pharmacol 2017; 8:667. [PMID: 29033838 PMCID: PMC5627017 DOI: 10.3389/fphar.2017.00667] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/07/2017] [Indexed: 11/13/2022] Open
Abstract
The pharmacodynamics of drug-candidates is often optimized by metrics that describe target binding (Kd or Ki value) or target modulation (IC50). However, these metrics are determined at equilibrium conditions, and consequently information regarding the onset and offset of target engagement and modulation is lost. Drug-target residence time is a measure for the lifetime of the drug-target complex, which has recently been receiving considerable interest, as target residence time is shown to have prognostic value for the in vivo efficacy of several drugs. In this study, we have investigated the relation between the increased residence time of antihistamines at the histamine H1 receptor (H1R) and the duration of effective target-inhibition by these antagonists. Hela cells, endogenously expressing low levels of the H1R, were incubated with a series of antihistamines and dissociation was initiated by washing away the unbound antihistamines. Using a calcium-sensitive fluorescent dye and a label free, dynamic mass redistribution based assay, functional recovery of the H1R responsiveness was measured by stimulating the cells with histamine over time, and the recovery was quantified as the receptor recovery time. Using these assays, we determined that the receptor recovery time for a set of antihistamines differed more than 40-fold and was highly correlated to their H1R residence times, as determined with competitive radioligand binding experiments to the H1R in a cell homogenate. Thus, the receptor recovery time is proposed as a cell-based and physiologically relevant metric for the lead optimization of G protein-coupled receptor antagonists, like the H1R antagonists. Both, label-free or real-time, classical signaling assays allow an efficient and physiologically relevant determination of kinetic properties of drug molecules.
Collapse
Affiliation(s)
- Reggie Bosma
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| | - Gesa Witt
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Lea A I Vaas
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Ivana Josimovic
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| | - Philip Gribbon
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Henry F Vischer
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Rob Leurs
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| |
Collapse
|
27
|
Bot I, Ortiz Zacarías NV, de Witte WEA, de Vries H, van Santbrink PJ, van der Velden D, Kröner MJ, van der Berg DJ, Stamos D, de Lange ECM, Kuiper J, IJzerman AP, Heitman LH. A novel CCR2 antagonist inhibits atherogenesis in apoE deficient mice by achieving high receptor occupancy. Sci Rep 2017; 7:52. [PMID: 28246398 PMCID: PMC5427923 DOI: 10.1038/s41598-017-00104-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 02/06/2017] [Indexed: 12/21/2022] Open
Abstract
CC Chemokine Receptor 2 (CCR2) and its endogenous ligand CCL2 are involved in a number of diseases, including atherosclerosis. Several CCR2 antagonists have been developed as potential therapeutic agents, however their in vivo clinical efficacy was limited. In this report, we aimed to determine whether 15a, an antagonist with a long residence time on the human CCR2, is effective in inhibiting the development of atherosclerosis in a mouse disease model. First, radioligand binding assays were performed to determine affinity and binding kinetics of 15a on murine CCR2. To assess the in vivo efficacy, western-type diet fed apoE-/- mice were treated daily with 15a or vehicle as control. Treatment with 15a reduced the amount of circulating CCR2+ monocytes and the size of the atherosclerotic plaques in both the carotid artery and the aortic root. We then showed that the long pharmacokinetic half-life of 15a combined with the high drug concentrations ensured prolonged CCR2 occupancy. These data render 15a a promising compound for drug development and confirms high receptor occupancy as a key parameter when targeting chemokine receptors.
Collapse
Affiliation(s)
- Ilze Bot
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | - Natalia V Ortiz Zacarías
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | - Wilhelmus E A de Witte
- Division of Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | - Henk de Vries
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | - Peter J van Santbrink
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | - Daniël van der Velden
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | - Mara J Kröner
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | - Dirk-Jan van der Berg
- Division of Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | | | - Elizabeth C M de Lange
- Division of Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | - Johan Kuiper
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | - Adriaan P IJzerman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | - Laura H Heitman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands.
| |
Collapse
|
28
|
Evolution of physicochemical properties of melanin concentrating hormone receptor 1 (MCHr1) antagonists. Bioorg Med Chem Lett 2016; 26:4559-4564. [PMID: 27595423 DOI: 10.1016/j.bmcl.2016.08.072] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/12/2016] [Accepted: 08/20/2016] [Indexed: 02/07/2023]
Abstract
One pharmacological principle for the treatment of obesity is blockade of the melanin concentrating hormone receptor 1 (MCHr1), which in rodents has been shown to be strongly associated with food intake and energy expenditure. However, discovery of safe and efficacious MCHr1 antagonists has proved to be complex. So far, six compounds have been progressed into clinical trials, but clinical validation of the concept is still lacking. An account of discovery of the three most recent clinical candidates targeting the MCHr1 receptor is given, with an emphasis on their physicochemical properties.
Collapse
|
29
|
Nederpelt I, Bleeker D, Tuijt B, IJzerman AP, Heitman LH. Kinetic binding and activation profiles of endogenous tachykinins targeting the NK1 receptor. Biochem Pharmacol 2016; 118:88-95. [PMID: 27501920 DOI: 10.1016/j.bcp.2016.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 08/02/2016] [Indexed: 02/01/2023]
Abstract
Ligand-receptor binding kinetics (i.e. association and dissociation rates) are emerging as important parameters for drug efficacy in vivo. Awareness of the kinetic behavior of endogenous ligands is pivotal, as drugs often have to compete with those. The binding kinetics of neurokinin 1 (NK1) receptor antagonists have been widely investigated while binding kinetics of endogenous tachykinins have hardly been reported, if at all. Therefore, the aim of this research was to investigate the binding kinetics of endogenous tachykinins and derivatives thereof and their role in the activation of the NK1 receptor. We determined the binding kinetics of seven tachykinins targeting the NK1 receptor. Dissociation rate constants (koff) ranged from 0.026±0.0029min-1 (Sar9,Met(O2)11-SP) to 0.21±0.015min-1 (septide). Association rate constants (kon) were more diverse: substance P (SP) associated the fastest with a kon value of 0.24±0.046nM-1min-1 while neurokinin A (NKA) had the slowest association rate constant of 0.001±0.0002nM-1min-1. Kinetic binding parameters were highly correlated with potency and maximal response values determined in label-free impedance-based experiments on U-251 MG cells. Our research demonstrates large variations in binding kinetics of tachykinins which correlate to receptor activation. These findings provide new insights into the ligand-receptor interactions of tachykinins and underline the importance of measuring binding kinetics of both drug candidates and competing endogenous ligands.
Collapse
Affiliation(s)
- I Nederpelt
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - D Bleeker
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - B Tuijt
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - A P IJzerman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - L H Heitman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands.
| |
Collapse
|
30
|
Nederpelt I, Vergroesen R, IJzerman A, Heitman L. Persistent GnRH receptor activation in pituitary αT3-1 cells analyzed with a label-free technology. Biosens Bioelectron 2016; 79:721-7. [DOI: 10.1016/j.bios.2015.12.066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/10/2015] [Accepted: 12/20/2015] [Indexed: 12/21/2022]
|
31
|
Guo D, Heitman LH, IJzerman AP. Kinetic Aspects of the Interaction between Ligand and G Protein-Coupled Receptor: The Case of the Adenosine Receptors. Chem Rev 2016; 117:38-66. [DOI: 10.1021/acs.chemrev.6b00025] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Dong Guo
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Laura H. Heitman
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Adriaan P. IJzerman
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| |
Collapse
|
32
|
de Witte WEA, Wong YC, Nederpelt I, Heitman LH, Danhof M, van der Graaf PH, Gilissen RAHJ, de Lange ECM. Mechanistic models enable the rational use of in vitro drug-target binding kinetics for better drug effects in patients. Expert Opin Drug Discov 2015; 11:45-63. [PMID: 26484747 DOI: 10.1517/17460441.2016.1100163] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Drug-target binding kinetics are major determinants of the time course of drug action for several drugs, as clearly described for the irreversible binders omeprazole and aspirin. This supports the increasing interest to incorporate newly developed high-throughput assays for drug-target binding kinetics in drug discovery. A meaningful application of in vitro drug-target binding kinetics in drug discovery requires insight into the relation between in vivo drug effect and in vitro measured drug-target binding kinetics. AREAS COVERED In this review, the authors discuss both the relation between in vitro and in vivo measured binding kinetics and the relation between in vivo binding kinetics, target occupancy and effect profiles. EXPERT OPINION More scientific evidence is required for the rational selection and development of drug-candidates on the basis of in vitro estimates of drug-target binding kinetics. To elucidate the value of in vitro binding kinetics measurements, it is necessary to obtain information on system-specific properties which influence the kinetics of target occupancy and drug effect. Mathematical integration of this information enables the identification of drug-specific properties which lead to optimal target occupancy and drug effect in patients.
Collapse
Affiliation(s)
- Wilhelmus E A de Witte
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Yin Cheong Wong
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Indira Nederpelt
- b Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Laura H Heitman
- b Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Meindert Danhof
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Piet H van der Graaf
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Ron A H J Gilissen
- c A Division of Janssen Pharmaceutica N.V., Janssen Research and Development , Turnhoutseweg 30, Beerse 2340 , Belgium
| | - Elizabeth C M de Lange
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| |
Collapse
|
33
|
Murphy DJ, Ou Y, Euler DH, Wessner K, Adamski S, Luo B, Wesolowski GA, Vogel R, Glantschnig H, Lubbers LS, Carroll SS, Lai MT. Determination of in Vivo Enzyme Occupancy Utilizing Inhibitor Dissociation Kinetics. J Am Chem Soc 2015; 137:11230-3. [PMID: 26301491 DOI: 10.1021/jacs.5b06518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
During drug discovery, assessment of in vivo target occupancy by therapeutic candidates is often required for predicting clinical efficacy. Current strategies for determining target occupancy include using radiolabeled or irreversible surrogates, which can be technically challenging, and the results are often not sufficiently quantitative. We developed a straightforward method by applying slow-dissociation kinetics to quantitatively determine enzyme occupancy without using specialized reagents. We applied this method to determine occupancy of Cathepsin K inhibitors in bone tissues harvested from rabbit femurs. Tissues from dosed animals were harvested, flash frozen, lysed, then analyzed by a jump-dilution assay with substrate. The rate of substrate turnover was monitored continuously until reaching steady state and progress curves were fit with the equation [product] = vst + ((vi - vs)/kobs)(1 - exp(-kobst)). The initial rate vi represents the residual activity of the enzyme before inhibitor dissociation; vs is the reaction rate after dissociation of the inhibitor. Occupancy is derived from the ratio of vi/vs. A significant benefit of the method is that data from both the occupied and unoccupied states are obtained in the same assay under identical conditions, which provides greater consistency between studies. The Cat K inhibitor MK-0674 (in vitro IC50 1 nM) was tested in young rabbits (<6 month old) and showed a dose-dependent increase in occupancy, reaching essentially complete occupancy at 1.0 mg/kg. In addition the method enables measurement of the total Cat K in the target tissue. Results confirmed complete occupancy even as the osteoclasts responded to higher doses with increased enzyme production.
Collapse
Affiliation(s)
- Dennis J Murphy
- In Vitro Pharmacology, ‡Bone Biology, and §In Vivo Pharmacology, Merck Research Laboratories , West Point, Pennsylvania 19486, United States
| | - Yangsi Ou
- In Vitro Pharmacology, ‡Bone Biology, and §In Vivo Pharmacology, Merck Research Laboratories , West Point, Pennsylvania 19486, United States
| | - Danielle H Euler
- In Vitro Pharmacology, ‡Bone Biology, and §In Vivo Pharmacology, Merck Research Laboratories , West Point, Pennsylvania 19486, United States
| | - Keith Wessner
- In Vitro Pharmacology, ‡Bone Biology, and §In Vivo Pharmacology, Merck Research Laboratories , West Point, Pennsylvania 19486, United States
| | - Sharon Adamski
- In Vitro Pharmacology, ‡Bone Biology, and §In Vivo Pharmacology, Merck Research Laboratories , West Point, Pennsylvania 19486, United States
| | - Bin Luo
- In Vitro Pharmacology, ‡Bone Biology, and §In Vivo Pharmacology, Merck Research Laboratories , West Point, Pennsylvania 19486, United States
| | - Gregg A Wesolowski
- In Vitro Pharmacology, ‡Bone Biology, and §In Vivo Pharmacology, Merck Research Laboratories , West Point, Pennsylvania 19486, United States
| | - Robert Vogel
- In Vitro Pharmacology, ‡Bone Biology, and §In Vivo Pharmacology, Merck Research Laboratories , West Point, Pennsylvania 19486, United States
| | - Helmut Glantschnig
- In Vitro Pharmacology, ‡Bone Biology, and §In Vivo Pharmacology, Merck Research Laboratories , West Point, Pennsylvania 19486, United States
| | - Laura S Lubbers
- In Vitro Pharmacology, ‡Bone Biology, and §In Vivo Pharmacology, Merck Research Laboratories , West Point, Pennsylvania 19486, United States
| | - Stephen S Carroll
- In Vitro Pharmacology, ‡Bone Biology, and §In Vivo Pharmacology, Merck Research Laboratories , West Point, Pennsylvania 19486, United States
| | - Ming-Tain Lai
- In Vitro Pharmacology, ‡Bone Biology, and §In Vivo Pharmacology, Merck Research Laboratories , West Point, Pennsylvania 19486, United States
| |
Collapse
|
34
|
Guo D, Heitman LH, IJzerman AP. The Role of Target Binding Kinetics in Drug Discovery. ChemMedChem 2015; 10:1793-6. [DOI: 10.1002/cmdc.201500310] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Indexed: 12/31/2022]
Affiliation(s)
- Dong Guo
- Division of Medicinal Chemistry; Leiden Academic Centre for Drug Research (LACDR); P.O. Box 9502 2300 RA Leiden the Netherlands
| | - Laura H. Heitman
- Division of Medicinal Chemistry; Leiden Academic Centre for Drug Research (LACDR); P.O. Box 9502 2300 RA Leiden the Netherlands
| | - Adriaan P. IJzerman
- Division of Medicinal Chemistry; Leiden Academic Centre for Drug Research (LACDR); P.O. Box 9502 2300 RA Leiden the Netherlands
| |
Collapse
|
35
|
Ruzza C, Rizzi A, Malfacini D, Molinari S, Giuliano C, Lovati E, Pietra C, Calo' G. In vitro and in vivo pharmacological characterization of Pronetupitant, a prodrug of the neurokinin 1 receptor antagonist Netupitant. Peptides 2015; 69:26-32. [PMID: 25843024 DOI: 10.1016/j.peptides.2015.03.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/18/2015] [Accepted: 03/26/2015] [Indexed: 10/23/2022]
Abstract
The aim of the present study was to investigate the pharmacological activity of Pronetupitant, a novel compound designed to act as prodrug of the NK1 antagonist Netupitant. In receptor binding experiments Pronetupitant displayed high selectivity for the NK1 receptor. In a calcium mobilization assay performed on CHONK1 cells Pronetupitant (100 nM, 15 min preincubation) behaved as an NK1 antagonist more potent than Netupitant (pK(B) 8.72 and 7.54, respectively). In the guinea pig ileum bioassay Pronetupitant antagonized the contractile effect of SP showing a similar potency as Netupitant (pK(B)≈9). Similar results were obtained with 5 min preincubation time while at 2 min only Pronetupitant produced significant effects. In vivo in mice the intrathecal injection of 0.1 nmol SP elicited the typical scratching, biting and licking (SBL) nociceptive response. This effect of SP was dose dependently (0.1-10 mg/kg) antagonized by Pronetupitant given intravenously 2 h before the peptide. Superimposable results were obtained using Netupitant. Pharmacokinetic studies performed in rats demonstrate that Pronetupitant, after i.v. administration, is quickly (few minutes) and completely converted to Netupitant. Collectively the present results indicated that Pronetupitant acts in vitro as selective NK1 antagonist more potent than Netupitant. However based on the short half-life measured for Pronetupitant in rats, the in vivo action of Pronetupitant can be entirely interpreted as due to its conversion to Netupitant.
Collapse
Affiliation(s)
- Chiara Ruzza
- Department of Medical Sciences, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, 44121 Ferrara, Italy
| | - Anna Rizzi
- Department of Medical Sciences, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, 44121 Ferrara, Italy
| | - Davide Malfacini
- Department of Medical Sciences, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, 44121 Ferrara, Italy
| | - Stefano Molinari
- Department of Medical Sciences, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, 44121 Ferrara, Italy
| | - Claudio Giuliano
- Preclinical Research and Development Department, Helsinn Healthcare SA, Lugano, Switzerland
| | - Emanuela Lovati
- Preclinical Research and Development Department, Helsinn Healthcare SA, Lugano, Switzerland
| | - Claudio Pietra
- Preclinical Research and Development Department, Helsinn Healthcare SA, Lugano, Switzerland
| | - Girolamo Calo'
- Department of Medical Sciences, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
36
|
Design strategies to address kinetics of drug binding and residence time. Bioorg Med Chem Lett 2015; 25:2019-27. [DOI: 10.1016/j.bmcl.2015.02.027] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 02/06/2015] [Accepted: 02/11/2015] [Indexed: 02/06/2023]
|
37
|
In vitro comparison of duration of action of melatonin agonists on melatonin MT1 receptor: possible link between duration of action and dissociation rate from receptor. Eur J Pharmacol 2015; 757:42-52. [PMID: 25797281 DOI: 10.1016/j.ejphar.2015.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 02/03/2015] [Accepted: 03/01/2015] [Indexed: 12/15/2022]
Abstract
Melatonin MT1 and MT2 receptors are Gi protein-coupled receptors and promising therapeutic targets for a number of diseases. A proportion of G protein-coupled receptor agonists and antagonists have been classified according to their duration of action, which influences their pharmacological efficacy. However, the duration of action of melatonin agonists remains unclear. In this study, we investigated the duration of action of melatonin agonists (melatonin, 2-iodomelatonin, ramelteon, and the ramelteon metabolite M-II) at the melatonin MT1 receptor, which is more resistant to agonist-induced desensitization than the melatonin MT2 receptor. In Chinese hamster ovary cells stably expressing the human melatonin MT1 receptor, significant differences in the duration of action were observed after 2-h pretreatment with agonists followed by washout. In contrast to melatonin and M-II, the agonist activities of ramelteon and 2-iodomelatonin were persistent (i.e. inhibition of forskolin-stimulated cAMP formation and increase in ERK 1/2 phosphorylation) even after repeated washouts. Similar activities were observed for INS-1 cells endogenously expressing the rat MT1 receptor. Further, we examined potential factors linked to the duration of action. Residual activities of melatonin agonists after washout strongly correlated with their dissociation rates from the human melatonin MT1 receptor, but not their lipophilicity or extent of desensitization. These data suggest that the in vitro duration of action significantly differs between melatonin agonists and might dictate dissociation kinetics. Characterization of these in vitro properties may facilitate further in vivo study of the duration of action.
Collapse
|
38
|
Endo T, Saijo T, Haneda E, Maeda J, Tokunaga M, Zhang MR, Kannami A, Asai H, Suzuki M, Suhara T, Higuchi M. Quantification of central substance P receptor occupancy by aprepitant using small animal positron emission tomography. Int J Neuropsychopharmacol 2015; 18:pyu030. [PMID: 25609595 PMCID: PMC4368889 DOI: 10.1093/ijnp/pyu030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Central substance P receptors, termed NK-1 receptors, have been considered as therapeutic targets in the development of drugs against diverse conditions, including emesis, overactive bladder, and depression. METHODS Here, we applied small animal positron emission tomography (PET) and a radioligand for NK-1 receptors ([(18)F]FE-SPA-RQ) for measuring occupancies of these receptors by a selective antagonist (aprepitant) in order to examine the validity of this in vivo imaging system for preclinical characterization of candidate agents acting on NK-1 receptors, and as a tool for predicting optimal doses in humans. RESULTS PET in gerbils depicted high uptake in the striatum and dose-dependent displacement with increasing doses of aprepitant. Occupancies increased as a function of aprepitant plasma concentrations according to a one-site competition model, which agrees with reported occupancy-concentration relationships in clinical studies after correction for species differences in plasma protein-unbound aprepitant fractions. These occupancy data were further supported by ex vivo autoradiography of brain samples from aprepitant-treated gerbils. In a pilot study of a marmoset, we obtained more accurate determinations of NK-1 receptor occupancy, less affected by spillover of signals from extracranial tissues than in gerbil experiments. CONCLUSIONS These findings support the utility of small animals and quantitative PET in the development of drugs targeting NK-1 receptors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Makoto Higuchi
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan (Drs Endo, Saijo, Haneda, Maeda, Tokunaga, Zhang, Suhara, Higuchi); Department of Molecular Neuroimaging, Tohoku University Graduate School of Medicine, Sendai, Japan (Drs Endo, Saijo, Suhara, and Higuchi); DMPK Research Laboratory, Mitsubishi Tanabe Pharma Corporation, Kisarazu, Japan (Drs Endo, Saijo, and Kannami); Clinical & Research Quality Assurance Department, Mitsubishi Tanabe Pharma Corporation, Tokyo, Japan (Dr Asai); Clinical Pharmacology Department, Mitsubishi Tanabe Pharma Corporation, Tokyo, Japan (Dr Suzuki).
| |
Collapse
|
39
|
Doyle JR, Harwood BN, Krishnaji ST, Krishnamurthy VM, Lin WE, Fortin JP, Kumar K, Kopin AS. A two-step strategy to enhance activity of low potency peptides. PLoS One 2014; 9:e110502. [PMID: 25391026 PMCID: PMC4229100 DOI: 10.1371/journal.pone.0110502] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 09/15/2014] [Indexed: 12/25/2022] Open
Abstract
Novel strategies are needed to expedite the generation and optimization of peptide probes targeting G protein-coupled receptors (GPCRs). We have previously shown that membrane tethered ligands (MTLs), recombinant proteins comprised of a membrane anchor, an extracellular linker, and a peptide ligand can be used to identify targeted receptor modulators. Although MTLs provide a useful tool to identify and/or modify functionally active peptides, a major limitation of this strategy is the reliance on recombinant protein expression. We now report the generation and pharmacological characterization of prototype peptide-linker-lipid conjugates, synthetic membrane anchored ligands (SMALs), which are designed as mimics of corresponding MTLs. In this study, we systematically compare the activity of selected peptides as MTLs versus SMALs. As prototypes, we focused on the precursor proteins of mature Substance P (SubP) and Cholecystokinin 4 (CCK4), specifically non-amidated SubP (SubP-COOH) and glycine extended CCK4 (CCK4-Gly-COOH). As low affinity soluble peptides these ligands each presented a challenging test case for assessment of MTL/SMAL technology. For each ligand, MTLs and corresponding SMALs showed agonist activity and comparable subtype selectivity. In addition, our results illustrate that membrane anchoring increases ligand potency. Furthermore, both MTL and SMAL induced signaling can be blocked by specific non-peptide antagonists suggesting that the anchored constructs may be orthosteric agonists. In conclusion, MTLs offer a streamlined approach for identifying low activity peptides which can be readily converted to higher potency SMALs. The ability to recapitulate MTL activity with SMALs extends the utility of anchored peptides as probes of GPCR function.
Collapse
Affiliation(s)
- Jamie R. Doyle
- Tufts Medical Center, Molecular Cardiology Research Institute, Molecular Pharmacology Research Center, Boston, Massachusetts, United States of America
| | - Benjamin N. Harwood
- Tufts Medical Center, Molecular Cardiology Research Institute, Molecular Pharmacology Research Center, Boston, Massachusetts, United States of America
- Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts, United States of America
| | | | - Vijay M. Krishnamurthy
- Tufts University, Department of Chemistry, Medford, Massachusetts, United States of America
| | - Wei-En Lin
- Tufts University, Department of Chemistry, Medford, Massachusetts, United States of America
| | - Jean-Philippe Fortin
- Tufts Medical Center, Molecular Cardiology Research Institute, Molecular Pharmacology Research Center, Boston, Massachusetts, United States of America
| | - Krishna Kumar
- Tufts University, Department of Chemistry, Medford, Massachusetts, United States of America
| | - Alan S. Kopin
- Tufts Medical Center, Molecular Cardiology Research Institute, Molecular Pharmacology Research Center, Boston, Massachusetts, United States of America
- Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts, United States of America
| |
Collapse
|
40
|
Mould R, Brown J, Marshall FH, Langmead CJ. Binding kinetics differentiates functional antagonism of orexin-2 receptor ligands. Br J Pharmacol 2014; 171:351-63. [PMID: 23692283 DOI: 10.1111/bph.12245] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 03/13/2013] [Accepted: 03/20/2013] [Indexed: 12/13/2022] Open
Abstract
Orexin receptor antagonism represents a novel approach for the treatment of insomnia that directly targets sleep/wake regulation. Several such compounds have entered into clinical development, including the dual orexin receptor antagonists, suvorexant and almorexant. In this study, we have used equilibrium and kinetic binding studies with the orexin-2 (OX₂) selective antagonist radioligand, [³H]-EMPA, to profile several orexin receptor antagonists. Furthermore, selected compounds were studied in cell-based assays of inositol phosphate accumulation and ERK-1/2 phosphorylation in CHO cells stably expressing the OX2 receptor that employ different agonist incubation times (30 and 5 min, respectively). EMPA, suvorexant, almorexant and TCS-OX-29 all bind to the OX₂ receptor with moderate to high affinity (pk(I) values ≥ 7.5), whereas the primarily OX1 selective antagonists SB-334867 and SB-408124 displayed low affinity (pK(I) values ca. 6). Competition kinetic analysis showed that the compounds displayed a range of dissociation rates from very fast (TCS-OX2-29, k(off) = 0.22 min⁻¹) to very slow (almorexant, k(off) = 0.005 min⁻¹). Notably, there was a clear correlation between association rate and affinity. In the cell-based assays, fast-offset antagonists EMPA and TCS-OX2-29 displayed surmountable antagonism of orexin-A agonist activity. However, both suvorexant and particularly almorexant cause concentration-dependent depression in the maximal orexin-A response, a profile that is more evident with a shorter agonist incubation time. Analysis according to a hemi-equilibrium model suggests that antagonist dissociation is slower in a cellular system than in membrane binding; under these conditions, almorexant effectively acts as a pseudo-irreversible antagonist.
Collapse
Affiliation(s)
- R Mould
- Heptares Therapeutics Ltd, BioPark, Broadwater Road, Welwyn Garden City, Herts, AL7 3AX, UK
| | | | | | | |
Collapse
|
41
|
Steinhoff MS, von Mentzer B, Geppetti P, Pothoulakis C, Bunnett NW. Tachykinins and their receptors: contributions to physiological control and the mechanisms of disease. Physiol Rev 2014; 94:265-301. [PMID: 24382888 DOI: 10.1152/physrev.00031.2013] [Citation(s) in RCA: 457] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The tachykinins, exemplified by substance P, are one of the most intensively studied neuropeptide families. They comprise a series of structurally related peptides that derive from alternate processing of three Tac genes and are expressed throughout the nervous and immune systems. Tachykinins interact with three neurokinin G protein-coupled receptors. The signaling, trafficking, and regulation of neurokinin receptors have also been topics of intense study. Tachykinins participate in important physiological processes in the nervous, immune, gastrointestinal, respiratory, urogenital, and dermal systems, including inflammation, nociception, smooth muscle contractility, epithelial secretion, and proliferation. They contribute to multiple diseases processes, including acute and chronic inflammation and pain, fibrosis, affective and addictive disorders, functional disorders of the intestine and urinary bladder, infection, and cancer. Neurokinin receptor antagonists are selective, potent, and show efficacy in models of disease. In clinical trials there is a singular success: neurokinin 1 receptor antagonists to treat nausea and vomiting. New information about the involvement of tachykinins in infection, fibrosis, and pruritus justifies further trials. A deeper understanding of disease mechanisms is required for the development of more predictive experimental models, and for the design and interpretation of clinical trials. Knowledge of neurokinin receptor structure, and the development of targeting strategies to disrupt disease-relevant subcellular signaling of neurokinin receptors, may refine the next generation of neurokinin receptor antagonists.
Collapse
|
42
|
Guo D, Hillger JM, IJzerman AP, Heitman LH. Drug-Target Residence Time-A Case for G Protein-Coupled Receptors. Med Res Rev 2014; 34:856-92. [DOI: 10.1002/med.21307] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Dong Guo
- Division of Medicinal Chemistry; Leiden Academic Centre for Drug Research; Leiden University; P.O. Box 9502 2300 RA Leiden the Netherlands
| | - Julia M. Hillger
- Division of Medicinal Chemistry; Leiden Academic Centre for Drug Research; Leiden University; P.O. Box 9502 2300 RA Leiden the Netherlands
| | - Adriaan P. IJzerman
- Division of Medicinal Chemistry; Leiden Academic Centre for Drug Research; Leiden University; P.O. Box 9502 2300 RA Leiden the Netherlands
| | - Laura H. Heitman
- Division of Medicinal Chemistry; Leiden Academic Centre for Drug Research; Leiden University; P.O. Box 9502 2300 RA Leiden the Netherlands
| |
Collapse
|
43
|
Camarda V, Ruzza C, Rizzi A, Trapella C, Guerrini R, Reinscheid RK, Calo G. In vitro and in vivo pharmacological characterization of the novel neuropeptide S receptor ligands QA1 and PI1. Peptides 2013; 48:27-35. [PMID: 23911665 DOI: 10.1016/j.peptides.2013.07.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 07/17/2013] [Accepted: 07/17/2013] [Indexed: 11/22/2022]
Abstract
The pharmacological activity of the novel neuropeptide S (NPS) receptor (NPSR) ligands QA1 and PI1 was investigated. In vitro QA1 and PI1 were tested in calcium mobilization studies performed in HEK293 cells expressing the recombinant mouse (HEK293mNPSR) and human (HEK293hNPSRIle107 and HEK293hNPSRAsn107) NPSR receptors. In vivo the compounds were studied in mouse righting reflex (RR) and locomotor activity (LA) tests. NPS caused a concentration dependent mobilization of intracellular calcium in the three cell lines with high potency (pEC50 8.73-9.14). In inhibition response curve and Schild protocol experiments the effects of NPS were antagonized by QA1 and PI1. QA1 displayed high potency (pKB 9.60-9.82) behaving as a insurmountable antagonist. However in coinjection experiments QA1 produced a rightward swift of the concentration response curve to NPS without modifying its maximal effects; this suggests that QA1 is actually a slow dissociating competitive antagonist. PI1 displayed a competitive type of antagonism and lower values of potencies (pA2 7.74-8.45). In vivo in mice NPS (0.1 nmol, i.c.v.) elicited arousal promoting action in the RR assay and stimulant effects in the LA test. QA1 (30 mgkg(-1)) was able to partially counteract the arousal promoting NPS effects, while PI1 was inactive in the RR test. In the LA test QA1 and PI1 only poorly blocked the NPS stimulant action. The present data demonstrated that QA1 and PI1 act as potent NPSR antagonists in vitro, however their usefulness for in vivo investigations in mice seems limited probably by pharmacokinetic reasons.
Collapse
Affiliation(s)
- V Camarda
- Department of Medical Science, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, 44121 Ferrara, Italy
| | | | | | | | | | | | | |
Collapse
|
44
|
Copeland RA. The dynamics of drug-target interactions: drug-target residence time and its impact on efficacy and safety. Expert Opin Drug Discov 2012; 5:305-10. [PMID: 22823083 DOI: 10.1517/17460441003677725] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The extent and duration of pharmacological action is determined by the lifetime of drug occupancy on a molecular target. This lifetime is defined by dynamic processes that control the rates of drug association and dissociation from the target. Recently, the term residence time has been coined to describe experimental measurements that can be related to the lifetime of the binary drug-target complex, and this in turn to durable, pharmacodynamic activity. The residence time concept and its impact on drug optimization are reviewed here. Examples are provided that demonstrate how a long residence time can improve drug efficacy in vivo. Additionally, optimization of drug-target residence time can help to mitigate off-target mediated toxicity, hence, improving drug safety and tolerability. Recent applications of the residence time concept to both drug discovery and development are also presented.
Collapse
Affiliation(s)
- Robert A Copeland
- Epizyme, Inc., 840 Memorial Drive, Cambridge, MA 02139, USA 617-401-8261 ; 617-349-0707 ;
| |
Collapse
|
45
|
Faedo S, Perdonà E, Antolini M, di Fabio R, Merlo Pich E, Corsi M. Functional and binding kinetic studies make a distinction between OX1 and OX2 orexin receptor antagonists. Eur J Pharmacol 2012; 692:1-9. [DOI: 10.1016/j.ejphar.2012.07.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 06/21/2012] [Accepted: 07/02/2012] [Indexed: 10/28/2022]
|
46
|
Rizzi A, Campi B, Camarda V, Molinari S, Cantoreggi S, Regoli D, Pietra C, Calo' G. In vitro and in vivo pharmacological characterization of the novel NK₁ receptor selective antagonist Netupitant. Peptides 2012; 37:86-97. [PMID: 22732666 DOI: 10.1016/j.peptides.2012.06.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 06/18/2012] [Accepted: 06/18/2012] [Indexed: 11/24/2022]
Abstract
The novel NK(1) receptor ligand Netupitant has been characterized in vitro and in vivo. In calcium mobilization studies CHO cells expressing the human NK receptors responded to a panel of agonists with the expected order of potency. In CHO NK(1) cells Netupitant concentration-dependently antagonized the stimulatory effects of substance P (SP) showing insurmountable antagonism (pK(B) 8.87). In cells expressing NK(2) or NK(3) receptors Netupitant was inactive. In the guinea pig ileum Netupitant concentration-dependently depressed the maximal response to SP (pK(B) 7.85) and, in functional washout experiments, displayed persistent (up to 5h) antagonist effects. In mice the intrathecal injection of SP elicited the typical scratching, biting and licking response that was dose-dependently inhibited by Netupitant given intraperitoneally in the 1-10mg/kg dose range. In gerbils, foot tapping behavior evoked by the intracerebroventricular injection of a NK(1) agonist was dose-dependently counteracted by Netupitant given intraperitoneally (ID(50) 1.5mg/kg) or orally (ID(50) 0.5mg/kg). In time course experiments in gerbils Netupitant displayed long lasting effects. In all the assays Aprepitant elicited similar effects as Netupitant. These results suggest that Netupitant behaves as a brain penetrant, orally active, potent and selective NK(1) antagonist. Thus this molecule can be useful for investigating the NK(1) receptor role in the control of central and peripheral functions. Netupitant has clinical potential in conditions such as chemotherapy induced nausea and vomiting, in which the blockade of NK(1) receptors has been demonstrated valuable for patients.
Collapse
Affiliation(s)
- Anna Rizzi
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, 44100 Ferrara, Italy
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Bissantz C, Bohnert C, Hoffmann T, Marcuz A, Schnider P, Malherbe P. Identification of a Crucial Amino Acid in the Helix Position 6.51 of Human Tachykinin Neurokinin 1 and 3 Receptors Contributing to the Insurmountable Mode of Antagonism by Dual NK1/NK3 Antagonists. J Med Chem 2012; 55:5061-76. [DOI: 10.1021/jm2017072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Caterina Bissantz
- Medicinal Chemistry and ‡DTA CNS, pRED, Pharma Research and Early Development, F. Hoffmann-La Roche AG, Grenzacherstrasse 124, CH4070, Basel, Switzerland
| | - Claudia Bohnert
- Medicinal Chemistry and ‡DTA CNS, pRED, Pharma Research and Early Development, F. Hoffmann-La Roche AG, Grenzacherstrasse 124, CH4070, Basel, Switzerland
| | - Torsten Hoffmann
- Medicinal Chemistry and ‡DTA CNS, pRED, Pharma Research and Early Development, F. Hoffmann-La Roche AG, Grenzacherstrasse 124, CH4070, Basel, Switzerland
| | - Anne Marcuz
- Medicinal Chemistry and ‡DTA CNS, pRED, Pharma Research and Early Development, F. Hoffmann-La Roche AG, Grenzacherstrasse 124, CH4070, Basel, Switzerland
| | - Patrick Schnider
- Medicinal Chemistry and ‡DTA CNS, pRED, Pharma Research and Early Development, F. Hoffmann-La Roche AG, Grenzacherstrasse 124, CH4070, Basel, Switzerland
| | - Pari Malherbe
- Medicinal Chemistry and ‡DTA CNS, pRED, Pharma Research and Early Development, F. Hoffmann-La Roche AG, Grenzacherstrasse 124, CH4070, Basel, Switzerland
| |
Collapse
|
48
|
Gossas T, Vrang L, Henderson I, Sedig S, Sahlberg C, Lindström E, Danielson UH. Aliskiren displays long-lasting interactions with human renin. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2011; 385:219-24. [PMID: 22193701 DOI: 10.1007/s00210-011-0718-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Accepted: 12/05/2011] [Indexed: 10/14/2022]
Abstract
Aliskiren is a selective renin inhibitor recently approved for use in hypertension. Efficacy duration appears longer than what would be expected based on its circulating half-life. The aim was therefore to characterize the kinetics of the interaction between aliskiren and renin. The interaction was evaluated in three assays and compared with two other renin inhibitors including remikiren. First, the inhibition of recombinant human renin was assessed by monitoring the cleavage of fluorescent substrate. Second, human plasma renin activity (PRA) was monitored by measuring generated angiotensin I over 1 h in the presence or absence of inhibitor. Finally, the affinity, association and dissociation rate constants were determined by using a surface plasmon resonance (SPR) biosensor assay. Aliskiren and remikiren were found to be equipotent inhibitors of recombinant renin activity (K(i) ≤ 0.04 nM) while compound 1 displayed a K (i) value of 1 nM. PRA was efficiently inhibited by both aliskiren and remikiren with IC₅₀ values of 0.2-0.3 nM. Remikiren and aliskiren also displayed long-lasting interactions with immobilized renin having k (off) values of 0.18 and 0.11 × 10⁻³ s⁻¹ respectively. These dissociation rate constants corresponded to residence times of 1.5 and 2.5 h, respectively, while compound 1 had a residence time lasting only 3 min. It is therefore concluded that the long-lasting interaction between aliskiren and human renin may contribute to the 24 h anti-hypertensive effect seen in clinical trials and possibly also to target-mediated drug disposition.
Collapse
Affiliation(s)
- Thomas Gossas
- Department of Biochemistry and Organic Chemistry, BMC, Uppsala University, Box 576, 75123 Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
49
|
Catalani MP, Alvaro G, Bernasconi G, Bettini E, Bromidge SM, Heer J, Tedesco G, Tommasi S. Identification of novel NK1/NK3 dual antagonists for the potential treatment of schizophrenia. Bioorg Med Chem Lett 2011; 21:6899-904. [DOI: 10.1016/j.bmcl.2011.07.116] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Revised: 07/28/2011] [Accepted: 07/29/2011] [Indexed: 10/17/2022]
|
50
|
Miller DC, Klute W, Brown AD. Discovery of potent, metabolically stable purine CRF-1 antagonists with differentiated binding kinetic profiles. Bioorg Med Chem Lett 2011; 21:6108-11. [DOI: 10.1016/j.bmcl.2011.08.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 08/06/2011] [Accepted: 08/08/2011] [Indexed: 10/17/2022]
|