1
|
Sun S, Chowdhury S, Hemeon I, Hasan A, Wilson MS, Bergeron P, Jia Q, Zenova AY, Grimwood ME, Gong W, Decker SM, Bichler P, Andrez JC, Focken T, Ngyuen T, Zhu J, White AD, Bankar G, Howard S, Chang E, Khakh K, Lin S, Dean R, Johnson JP, Chang JH, Hackos DH, McKerrall SJ, Sellers B, Ortwine DF, Cohen CJ, Safina BS, Sutherlin DP, Dehnhardt CM. Discovery of novel cyclopentane carboxylic acids as potent and selective inhibitors of Na V1.7. Bioorg Med Chem Lett 2025; 116:130033. [PMID: 39580005 DOI: 10.1016/j.bmcl.2024.130033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/28/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
Discovery efforts leading to the identification of cyclopentane carboxylic acid 31, a potent inhibitor of NaV1.7 that showed high selectivity over NaV1.5 and exhibited robust analgesic effects in an inherited erythromelalgia (IEM) transgenic mouse assay, are described herein. Key design elements that culminated in the discovery of 31 include exploration of proline substituents, replacement of the proline warhead with cyclopentane carboxylic acid, that led to significantly boosted NaV1.7 potency, and replacement of the metabolically labile adamantane motif with the 2,6-dichlorobenzyl substituted piperidine system, that addressed metabolic instability and led to a significant improvement in PK.
Collapse
Affiliation(s)
- Shaoyi Sun
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada.
| | - Sultan Chowdhury
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Ivan Hemeon
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Abid Hasan
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Michael S Wilson
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | | | - Qi Jia
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Alla Y Zenova
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Mike E Grimwood
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Wei Gong
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Shannon M Decker
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Paul Bichler
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | | | - Thilo Focken
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Theresa Ngyuen
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | - Jiuxiang Zhu
- Chempartner, Building No. 5, 998 Halei Rd., Zhangjiang Hi-Tech Park, Pudong New Area, Shanghai, PR China
| | - Andrew D White
- Chempartner, Building No. 5, 998 Halei Rd., Zhangjiang Hi-Tech Park, Pudong New Area, Shanghai, PR China
| | - Girish Bankar
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Sarah Howard
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | - Elaine Chang
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Kuldip Khakh
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Sophia Lin
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Richard Dean
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - J P Johnson
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Jae H Chang
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | - David H Hackos
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | | | - Ben Sellers
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | - Dan F Ortwine
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | - Charles J Cohen
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Brian S Safina
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | | | | |
Collapse
|
2
|
Yu X, Zhao X, Li L, Huang Y, Cui C, Hu Q, Xu H, Yin B, Chen X, Zhao D, Qiu Y, Hou Y. Recent advances in small molecule Nav 1.7 inhibitors for cancer pain management. Bioorg Chem 2024; 150:107605. [PMID: 38971095 DOI: 10.1016/j.bioorg.2024.107605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/22/2024] [Accepted: 06/28/2024] [Indexed: 07/08/2024]
Abstract
The dorsal root ganglion (DRG) is the primary neuron responsible for transmitting peripheral pain signals to the central nervous system and plays a crucial role in pain transduction. Modulation of DRG excitability is considered a viable approach for pain management. Neuronal excitability is intricately linked to the ion channels on the neurons. The small and medium-sized DRG neurons are chiefly engaged in pain conduction and have high levels of TTX-S sodium channels, with Nav1.7 accounting for approximately 80% of the current. Voltage-gated sodium channel (VGSC or Nav) blockers are vital targets for the management of central nervous system diseases, particularly chronic pain. VGSCs play a key role in controlling cellular excitability. Clinical research has shown that Nav1.7 plays a crucial role in pain sensation, and there is strong genetic evidence linking Nav1.7 and its encoding gene SCN9A gene to painful disorders in humans. Many studies have shown that Nav1.7 plays an important role in pain management. The role of Nav1.7 in pain signaling pathways makes it an attractive target for the potential development of new pain drugs. Meanwhile, understanding the architecture of Nav1.7 may help to develop the next generation of painkillers. This review provides updates on the recently reported molecular inhibitors targeting the Nav1.7 pathway, summarizes their structure-activity relationships (SARs), and discusses their therapeutic effects on painful diseases. Pharmaceutical chemists are working to improve the therapeutic index of Nav1.7 inhibitors, achieve better analgesic effects, and reduce side effects. We hope that this review will contribute to the development of novel Nav1.7 inhibitors as potential drugs.
Collapse
Affiliation(s)
- Xiaoquan Yu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Xingyi Zhao
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Lingjun Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Yufeng Huang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Chaoyang Cui
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Qiaoguan Hu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Haoyu Xu
- Yangtze River Pharmaceutical (Group) Co., Ltd., 1 South Yangtze River Road, Taizhou City, Jiangsu Province, 225321, China
| | - Bixi Yin
- Yangtze River Pharmaceutical Group Jiangsu Haici Biological Pharmaceutical Co., Ltd., 8 Taizhen Road, Medical New & Hi-tech Industrial Development Zone, Taizhou City, Jiangsu Province, 225321, China
| | - Xiao Chen
- Yangtze River Pharmaceutical Group Jiangsu Haici Biological Pharmaceutical Co., Ltd., 8 Taizhen Road, Medical New & Hi-tech Industrial Development Zone, Taizhou City, Jiangsu Province, 225321, China
| | - Dong Zhao
- Yangtze River Pharmaceutical Group Jiangsu Haici Biological Pharmaceutical Co., Ltd., 8 Taizhen Road, Medical New & Hi-tech Industrial Development Zone, Taizhou City, Jiangsu Province, 225321, China
| | - Yue Qiu
- Yangtze River Pharmaceutical Group Jiangsu Haici Biological Pharmaceutical Co., Ltd., 8 Taizhen Road, Medical New & Hi-tech Industrial Development Zone, Taizhou City, Jiangsu Province, 225321, China
| | - Yunlei Hou
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China.
| |
Collapse
|
3
|
He G, Zheng Y, Chang S, Wang L, Yang X, Hao H, Li J, Zhang X, Tian F, Liang X, Xu H, Wang P, Chen X, Cao Z, Fang S, Gao Z, Liu H. Discovery of Novel Pyrimidine-Based Derivatives as Nav1.2 Inhibitors with Efficacy in Mouse Models of Epilepsy. J Med Chem 2024. [PMID: 39037114 DOI: 10.1021/acs.jmedchem.4c00861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Dysfunction of voltage-gated sodium channel Nav1.2 causes various epileptic disorders, and inhibition of the channel has emerged as an attractive therapeutic strategy. However, currently available Nav1.2 inhibitors exhibit low potency and limited structural diversity. In this study, a novel series of pyrimidine-based derivatives with Nav1.2 inhibitory activity were designed, synthesized, and evaluated. Compounds 14 and 35 exhibited potent activity against Nav1.2, boasting IC50 values of 120 and 65 nM, respectively. Compound 14 displayed favorable pharmacokinetics (F = 43%) following intraperitoneal injection and excellent brain penetration potency (B/P = 3.6). Compounds 14 and 35 exhibited robust antiepileptic activities in the maximal electroshock test, with ED50 values of 3.2 and 11.1 mg/kg, respectively. Compound 35 also demonstrated potent antiepileptic activity in a 6 Hz (32 mA) model, with an ED50 value of 18.5 mg/kg. Overall, compounds 14 and 35 are promising leads for the development of new small-molecule therapeutics for epilepsy.
Collapse
Affiliation(s)
- Guoxue He
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yueming Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shunzhen Chang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Long Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Xiaohao Yang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haishuang Hao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jiyuan Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xian Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Fuyun Tian
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xuewu Liang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haiyan Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Pei Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xueqin Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zeyu Cao
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sui Fang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhaobing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hong Liu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
4
|
Amador R, Tahrioui A, Barreau M, Lesouhaitier O, Smietana M, Clavé G. N-Acylsulfonamide: a valuable moiety to design new sulfa drug analogues. RSC Med Chem 2023; 14:1567-1571. [PMID: 37593573 PMCID: PMC10429802 DOI: 10.1039/d3md00229b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/04/2023] [Indexed: 08/19/2023] Open
Abstract
Sulfonamides are the oldest class of antibiotics, discovered more than 80 years ago. They are still used today despite the appearance of drug resistance phenomena that limit their prescription. Since the discovery and use of the first sulfa drugs, many analogues have been synthesized in order to obtain new active molecules able to circumvent bacterial resistance. Structurally similar to sulfonamide, the N-acylsulfonamide group arouses interest in the field of medicinal chemistry due to specific physico-chemical properties. We report here the synthesis and antibacterial/antibiofilm activities of 18 sulfa drug analogues with an N-acylsulfonamide moiety. These derivatives were obtained efficiently by sulfo-click reactions between readily available thioacid and sulfonyl azide synthons.
Collapse
Affiliation(s)
- Romain Amador
- Institut des Biomolécules Max Mousseron, Université de Montpellier, CNRS, ENSCM 1919 route de Mende 34095 Montpellier France
| | - Ali Tahrioui
- Univ Rouen Normandie, Université Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-infectieuses (CBSA) UR 4312 F-76000 Rouen France
| | - Magalie Barreau
- Univ Rouen Normandie, Université Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-infectieuses (CBSA) UR 4312 F-76000 Rouen France
| | - Olivier Lesouhaitier
- Univ Rouen Normandie, Université Caen Normandie, Normandie Univ, Communication Bactérienne et Stratégies Anti-infectieuses (CBSA) UR 4312 F-76000 Rouen France
| | - Michael Smietana
- Institut des Biomolécules Max Mousseron, Université de Montpellier, CNRS, ENSCM 1919 route de Mende 34095 Montpellier France
| | - Guillaume Clavé
- Institut des Biomolécules Max Mousseron, Université de Montpellier, CNRS, ENSCM 1919 route de Mende 34095 Montpellier France
| |
Collapse
|
5
|
Zidar N, Tomašič T, Kikelj D, Durcik M, Tytgat J, Peigneur S, Rogers M, Haworth A, Kirby RW. New aryl and acylsulfonamides as state-dependent inhibitors of Na v1.3 voltage-gated sodium channel. Eur J Med Chem 2023; 258:115530. [PMID: 37329714 DOI: 10.1016/j.ejmech.2023.115530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/19/2023]
Abstract
Voltage-gated sodium channels (Navs) play an essential role in neurotransmission, and their dysfunction is often a cause of various neurological disorders. The Nav1.3 isoform is found in the CNS and upregulated after injury in the periphery, but its role in human physiology has not yet been fully elucidated. Reports suggest that selective Nav1.3 inhibitors could be used as novel therapeutics to treat pain or neurodevelopmental disorders. Few selective inhibitors of this channel are known in the literature. In this work, we report the discovery of a new series of aryl and acylsulfonamides as state-dependent inhibitors of Nav1.3 channels. Using a ligand-based 3D similarity search and subsequent hit optimization, we identified and prepared a series of 47 novel compounds and tested them on Nav1.3, Nav1.5, and a selected subset also on Nav1.7 channels in a QPatch patch-clamp electrophysiology assay. Eight compounds had an IC50 value of less than 1 μM against the Nav1.3 channel inactivated state, with one compound displaying an IC50 value of 20 nM, whereas activity against the inactivated state of the Nav1.5 channel and Nav1.7 channel was approximately 20-fold weaker. None of the compounds showed use-dependent inhibition of the cardiac isoform Nav1.5 at a concentration of 30 μM. Further selectivity testing of the most promising hits was measured using the two-electrode voltage-clamp method against the closed state of the Nav1.1-Nav1.8 channels, and compound 15b displayed small, yet selective, effects against the Nav1.3 channel, with no activity against the other isoforms. Additional selectivity testing of promising hits against the inactivated state of the Nav1.3, Nav1.7, and Nav1.8 channels revealed several compounds with robust and selective activity against the inactivated state of the Nav1.3 channel among the three isoforms tested. Moreover, the compounds were not cytotoxic at a concentration of 50 μM, as demonstrated by the assay in human HepG2 cells (hepatocellular carcinoma cells). The novel state-dependent inhibitors of Nav1.3 discovered in this work provide a valuable tool to better evaluate this channel as a potential drug target.
Collapse
Affiliation(s)
- Nace Zidar
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia.
| | - Tihomir Tomašič
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia
| | - Danijel Kikelj
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia
| | - Martina Durcik
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia
| | - Jan Tytgat
- University of Leuven (KU Leuven), Toxicology & Pharmacology, O&N2, PO Box 922, Herestraat 49, 3000, Leuven, Belgium
| | - Steve Peigneur
- University of Leuven (KU Leuven), Toxicology & Pharmacology, O&N2, PO Box 922, Herestraat 49, 3000, Leuven, Belgium
| | - Marc Rogers
- Metrion Biosciences Limited, Building 2, Granta Centre, Granta Park, Great Abington, Cambridge, CB21 6AL, UK
| | - Alexander Haworth
- Metrion Biosciences Limited, Building 2, Granta Centre, Granta Park, Great Abington, Cambridge, CB21 6AL, UK
| | - Robert W Kirby
- Metrion Biosciences Limited, Building 2, Granta Centre, Granta Park, Great Abington, Cambridge, CB21 6AL, UK
| |
Collapse
|
6
|
Ouyang X, Su M, Xue D, Dong L, Niu H, Li W, Liu Y, Wang K, Shao L. Design, synthesis, and biological evaluation of acyl sulfonamide derivatives with spiro cycles as Na V1.7 inhibitors for antinociception. Bioorg Med Chem 2023; 86:117290. [PMID: 37137269 DOI: 10.1016/j.bmc.2023.117290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/30/2023] [Accepted: 04/13/2023] [Indexed: 05/05/2023]
Abstract
Chronic pain, as an unmet medical need, severely impacts the quality of life. The voltage-gated sodium channel NaV1.7 preferentially expressed in sensory neurons of dorsal root ganglia (DRG) serves a promising target for pain therapy. Here, we report the design, synthesis, and evaluation of a series of acyl sulfonamide derivatives targeting Nav1.7 for their antinociceptive activities. Among the derivatives tested, the compound 36c was identified as a selective and potent NaV1.7 inhibitor in vitro and exhibited antinociceptive effects in vivo. The identification of 36c not only provides a new insight into the discovery of selective NaV1.7 inhibitors, but also may hold premise for pain therapy.
Collapse
Affiliation(s)
- Xiangshuo Ouyang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Min Su
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China
| | - Dengqi Xue
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Liying Dong
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China
| | - Heling Niu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China.
| | - Yani Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China.
| | - KeWei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China.
| | - Liming Shao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China; State Key Laboratory of Medical Neurobiology, Fudan University, No. 138 Yixueyuan Road, Shanghai 200032, China.
| |
Collapse
|
7
|
Amador R, Delpal A, Canard B, Vasseur JJ, Decroly E, Debart F, Clavé G, Smietana M. Facile access to 4'-( N-acylsulfonamide) modified nucleosides and evaluation of their inhibitory activity against SARS-CoV-2 RNA cap N7-guanine-methyltransferase nsp14. Org Biomol Chem 2022; 20:7582-7586. [PMID: 36156055 DOI: 10.1039/d2ob01569b] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
N-Acylsulfonamides possess an additional carbonyl function compared to their sulfonamide analogues. Due to their unique physico-chemical properties, interest in molecules containing the N-acylsulfonamide moiety and especially nucleoside derivatives is growing in the field of medicinal chemistry. The recent renewal of interest in antiviral drugs derived from nucleosides containing a sulfonamide function has led us to evaluate the therapeutic potential of N-acylsulfonamide analogues. While these compounds are usually obtained by a difficult acylation of sulfonamides, we report here the easy and efficient synthesis of 20 4'-(N-acylsulfonamide) adenosine derivatives via the sulfo-click reaction. The target compounds were obtained from thioacid and sulfonyl azide synthons in excellent yields and were evaluated as potential inhibitors of the SARS-CoV-2 RNA cap N7-guanine-methyltransferase nsp14.
Collapse
Affiliation(s)
- Romain Amador
- Institut des Biomolécules Max Mousseron, Université de Montpellier, CNRS, ENSCM, 1919 route de Mende, 34095 Montpellier, France.
| | - Adrien Delpal
- Architecture et Fonction des Macromolécules Biologiques, Université d'Aix-Marseille, CNRS, 163 Avenue de Luminy, Marseille, France
| | - Bruno Canard
- Architecture et Fonction des Macromolécules Biologiques, Université d'Aix-Marseille, CNRS, 163 Avenue de Luminy, Marseille, France
| | - Jean-Jacques Vasseur
- Institut des Biomolécules Max Mousseron, Université de Montpellier, CNRS, ENSCM, 1919 route de Mende, 34095 Montpellier, France.
| | - Etienne Decroly
- Architecture et Fonction des Macromolécules Biologiques, Université d'Aix-Marseille, CNRS, 163 Avenue de Luminy, Marseille, France
| | - Françoise Debart
- Institut des Biomolécules Max Mousseron, Université de Montpellier, CNRS, ENSCM, 1919 route de Mende, 34095 Montpellier, France.
| | - Guillaume Clavé
- Institut des Biomolécules Max Mousseron, Université de Montpellier, CNRS, ENSCM, 1919 route de Mende, 34095 Montpellier, France.
| | - Michael Smietana
- Institut des Biomolécules Max Mousseron, Université de Montpellier, CNRS, ENSCM, 1919 route de Mende, 34095 Montpellier, France.
| |
Collapse
|
8
|
Kitano Y, Shinozuka T. Inhibition of Na V1.7: the possibility of ideal analgesics. RSC Med Chem 2022; 13:895-920. [PMID: 36092147 PMCID: PMC9384491 DOI: 10.1039/d2md00081d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/25/2022] [Indexed: 08/03/2023] Open
Abstract
The selective inhibition of NaV1.7 is a promising strategy for developing novel analgesic agents with fewer adverse effects. Although the potent selective inhibition of NaV1.7 has been recently achieved, multiple NaV1.7 inhibitors failed in clinical development. In this review, the relationship between preclinical in vivo efficacy and NaV1.7 coverage among three types of voltage-gated sodium channel (VGSC) inhibitors, namely conventional VGSC inhibitors, sulphonamides and acyl sulphonamides, is discussed. By demonstrating the PK/PD discrepancy of preclinical studies versus in vivo models and clinical results, the potential reasons behind the disconnect between preclinical results and clinical outcomes are discussed together with strategies for developing ideal analgesic agents.
Collapse
Affiliation(s)
- Yutaka Kitano
- R&D Division, Daiichi Sankyo Co., Ltd. 1-2-58 Hiromachi Shinagawa-ku Tokyo 140-8710 Japan
| | - Tsuyoshi Shinozuka
- R&D Division, Daiichi Sankyo Co., Ltd. 1-2-58 Hiromachi Shinagawa-ku Tokyo 140-8710 Japan
| |
Collapse
|
9
|
Elleman AV, Du Bois J. Chemical and Biological Tools for the Study of Voltage-Gated Sodium Channels in Electrogenesis and Nociception. Chembiochem 2022; 23:e202100625. [PMID: 35315190 PMCID: PMC9359671 DOI: 10.1002/cbic.202100625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/22/2022] [Indexed: 12/17/2022]
Abstract
The malfunction and misregulation of voltage-gated sodium channels (NaV s) underlie in large part the electrical hyperexcitability characteristic of chronic inflammatory and neuropathic pain. NaV s are responsible for the initiation and propagation of electrical impulses (action potentials) in cells. Tissue and nerve injury alter the expression and localization of multiple NaV isoforms, including NaV 1.1, 1.3, and 1.6-1.9, resulting in aberrant action potential firing patterns. To better understand the role of NaV regulation, localization, and trafficking in electrogenesis and pain pathogenesis, a number of chemical and biological reagents for interrogating NaV function have been advanced. The development and application of such tools for understanding NaV physiology are the focus of this review.
Collapse
Affiliation(s)
- Anna V Elleman
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - J Du Bois
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
10
|
Nguyen PT, Yarov-Yarovoy V. Towards Structure-Guided Development of Pain Therapeutics Targeting Voltage-Gated Sodium Channels. Front Pharmacol 2022; 13:842032. [PMID: 35153801 PMCID: PMC8830516 DOI: 10.3389/fphar.2022.842032] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium (NaV) channels are critical molecular determinants of action potential generation and propagation in excitable cells. Normal NaV channel function disruption can affect physiological neuronal signaling and lead to increased sensitivity to pain, congenital indifference to pain, uncoordinated movement, seizures, or paralysis. Human genetic studies have identified human NaV1.7 (hNaV1.7), hNaV1.8, and hNaV1.9 channel subtypes as crucial players in pain signaling. The premise that subtype selective NaV inhibitors can reduce pain has been reinforced through intensive target validation and therapeutic development efforts. However, an ideal therapeutic has yet to emerge. This review is focused on recent progress, current challenges, and future opportunities to develop NaV channel targeting small molecules and peptides as non-addictive therapeutics to treat pain.
Collapse
Affiliation(s)
- Phuong T Nguyen
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States.,Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
11
|
Niu HL, Liu YN, Xue DQ, Dong LY, Liu HJ, Wang J, Zheng YL, Zou AR, Shao LM, Wang K. Inhibition of Nav1.7 channel by a novel blocker QLS-81 for alleviation of neuropathic pain. Acta Pharmacol Sin 2021; 42:1235-1247. [PMID: 34103689 PMCID: PMC8285378 DOI: 10.1038/s41401-021-00682-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 04/14/2021] [Indexed: 02/02/2023] Open
Abstract
Voltage-gated sodium channel Nav1.7 robustly expressed in peripheral nociceptive neurons has been considered as a therapeutic target for chronic pain, but there is no selective Nav1.7 inhibitor available for therapy of chronic pain. Ralfinamide has shown anti-nociceptive activity in animal models of inflammatory and neuropathic pain and is currently under phase III clinical trial for neuropathic pain. Based on ralfinamide, a novel small molecule (S)-2-((3-(4-((2-fluorobenzyl) oxy) phenyl) propyl) amino) propanamide (QLS-81) was synthesized. Here, we report the electrophysiological and pharmacodynamic characterization of QLS-81 as a Nav1.7 channel inhibitor with promising anti-nociceptive activity. In whole-cell recordings of HEK293 cells stably expressing Nav1.7, QLS-81 (IC50 at 3.5 ± 1.5 μM) was ten-fold more potent than its parent compound ralfinamide (37.1 ± 2.9 μM) in inhibiting Nav1.7 current. QLS-81 inhibition on Nav1.7 current was use-dependent. Application of QLS-81 (10 μM) caused a hyperpolarizing shift of the fast and slow inactivation of Nav1.7 channel about 7.9 mV and 26.6 mV, respectively, and also slowed down the channel fast and slow inactivation recovery. In dissociated mouse DRG neurons, QLS-81 (10 μM) inhibited native Nav current and suppressed depolarizing current pulse-elicited neuronal firing. Administration of QLS-81 (2, 5, 10 mg· kg-1· d-1, i.p.) in mice for 10 days dose-dependently alleviated spinal nerve injury-induced neuropathic pain and formalin-induced inflammatory pain. In addition, QLS-81 (10 μM) did not significantly affect ECG in guinea pig heart ex vivo; and administration of QLS-81 (10, 20 mg/kg, i.p.) in mice had no significant effect on spontaneous locomotor activity. Taken together, our results demonstrate that QLS-81, as a novel Nav1.7 inhibitor, is efficacious on chronic pain in mice, and it may hold developmental potential for pain therapy.
Collapse
Affiliation(s)
- He-Ling Niu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266073, China
| | - Ya-Ni Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266073, China.
| | - Deng-Qi Xue
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Li-Ying Dong
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266073, China
| | - Hui-Jie Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266073, China
| | - Jing Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266073, China
| | - Yi-Lin Zheng
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - An-Ruo Zou
- Institute of Innovative Drug, Qingdao University, Qingdao, 266021, China
| | - Li-Ming Shao
- School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - KeWei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266073, China.
- Institute of Innovative Drug, Qingdao University, Qingdao, 266021, China.
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Tamar, Hong Kong, China.
| |
Collapse
|
12
|
Roecker AJ, Layton ME, Pero JE, Kelly MJ, Greshock TJ, Kraus RL, Li Y, Klein R, Clements M, Daley C, Jovanovska A, Ballard JE, Wang D, Zhao F, Brunskill APJ, Peng X, Wang X, Sun H, Houghton AK, Burgey CS. Discovery of Arylsulfonamide Na v1.7 Inhibitors: IVIVC, MPO Methods, and Optimization of Selectivity Profile. ACS Med Chem Lett 2021; 12:1038-1049. [PMID: 34141090 PMCID: PMC8201757 DOI: 10.1021/acsmedchemlett.1c00218] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/26/2021] [Indexed: 01/13/2023] Open
Abstract
The voltage-gated sodium channel Nav1.7 continues to be a high-profile target for the treatment of various pain afflictions due to its strong human genetic validation. While isoform selective molecules have been discovered and advanced into the clinic, to date, this target has yet to bear fruit in the form of marketed therapeutics for the treatment of pain. Lead optimization efforts over the past decade have focused on selectivity over Nav1.5 due to its link to cardiac side effects as well as the translation of preclinical efficacy to man. Inhibition of Nav1.6 was recently reported to yield potential respiratory side effects preclinically, and this finding necessitated a modified target selectivity profile. Herein, we report the continued optimization of a novel series of arylsulfonamide Nav1.7 inhibitors to afford improved selectivity over Nav1.6 while maintaining rodent oral bioavailability through the use of a novel multiparameter optimization (MPO) paradigm. We also report in vitro-in vivo correlations from Nav1.7 electrophysiology protocols to preclinical models of efficacy to assist in projecting clinical doses. These efforts produced inhibitors such as compound 19 with potency against Nav1.7, selectivity over Nav1.5 and Nav1.6, and efficacy in behavioral models of pain in rodents as well as inhibition of rhesus olfactory response indicative of target modulation.
Collapse
Affiliation(s)
- Anthony J. Roecker
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Mark E. Layton
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Joseph E. Pero
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Michael J. Kelly
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Thomas J. Greshock
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Richard L. Kraus
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Yuxing Li
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Rebecca Klein
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Michelle Clements
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Christopher Daley
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Aneta Jovanovska
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Jeanine E. Ballard
- Pharmacokinetic,
Pharmacodynamics, and Drug Metabolism, Merck
& Co., Inc., West Point, Pennsylvania 19486, United States
| | - Deping Wang
- Computational
and Structural Chemistry, Merck & Co.,
Inc., West Point, Pennsylvania 19486, United States
| | - Fuqiang Zhao
- Translational
Imaging and Biomarkers, Merck & Co.,
Inc., West Point, Pennsylvania 19486, United States
| | - Andrew P. J. Brunskill
- Molecular
and Materials Characterization, Merck &
Co., Inc., Rahway, New Jersey 07065, United States
| | - Xuanjia Peng
- HitS
Unite, WuXi AppTec Co., Ltd. (Shanghai), Shanghai 200131, China
| | - Xiu Wang
- IDSU, WuXi AppTec
Co., Ltd. (Shanghai), Shanghai 200131, China
| | - Haiyan Sun
- IDSU, WuXi AppTec
Co., Ltd. (Shanghai), Shanghai 200131, China
| | - Andrea K. Houghton
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Christopher S. Burgey
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
13
|
Structural Pharmacology of Voltage-Gated Sodium Channels. J Mol Biol 2021; 433:166967. [PMID: 33794261 DOI: 10.1016/j.jmb.2021.166967] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 12/19/2022]
Abstract
Voltage-gated sodium (NaV) channels initiate and propagate action potentials in excitable tissues to mediate key physiological processes including heart contraction and nervous system function. Accordingly, NaV channels are major targets for drugs, toxins and disease-causing mutations. Recent breakthroughs in cryo-electron microscopy have led to the visualization of human NaV1.1, NaV1.2, NaV1.4, NaV1.5 and NaV1.7 channel subtypes at high-resolution. These landmark studies have greatly advanced our structural understanding of channel architecture, ion selectivity, voltage-sensing, electromechanical coupling, fast inactivation, and the molecular basis underlying NaV channelopathies. NaV channel structures have also been increasingly determined in complex with toxin and small molecule modulators that target either the pore module or voltage sensor domains. These structural studies have provided new insights into the mechanisms of pharmacological action and opportunities for subtype-selective NaV channel drug design. This review will highlight the structural pharmacology of human NaV channels as well as the potential use of engineered and chimeric channels in future drug discovery efforts.
Collapse
|
14
|
Safina BS, McKerrall SJ, Sun S, Chen CA, Chowdhury S, Jia Q, Li J, Zenova AY, Andrez JC, Bankar G, Bergeron P, Chang JH, Chang E, Chen J, Dean R, Decker SM, DiPasquale A, Focken T, Hemeon I, Khakh K, Kim A, Kwan R, Lindgren A, Lin S, Maher J, Mezeyova J, Misner D, Nelkenbrecher K, Pang J, Reese R, Shields SD, Sojo L, Sheng T, Verschoof H, Waldbrook M, Wilson MS, Xie Z, Young C, Zabka TS, Hackos DH, Ortwine DF, White AD, Johnson JP, Robinette CL, Dehnhardt CM, Cohen CJ, Sutherlin DP. Discovery of Acyl-sulfonamide Na v1.7 Inhibitors GDC-0276 and GDC-0310. J Med Chem 2021; 64:2953-2966. [PMID: 33682420 DOI: 10.1021/acs.jmedchem.1c00049] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nav1.7 is an extensively investigated target for pain with a strong genetic link in humans, yet in spite of this effort, it remains challenging to identify efficacious, selective, and safe inhibitors. Here, we disclose the discovery and preclinical profile of GDC-0276 (1) and GDC-0310 (2), selective Nav1.7 inhibitors that have completed Phase 1 trials. Our initial search focused on close-in analogues to early compound 3. This resulted in the discovery of GDC-0276 (1), which possessed improved metabolic stability and an acceptable overall pharmacokinetics profile. To further derisk the predicted human pharmacokinetics and enable QD dosing, additional optimization of the scaffold was conducted, resulting in the discovery of a novel series of N-benzyl piperidine Nav1.7 inhibitors. Improvement of the metabolic stability by blocking the labile benzylic position led to the discovery of GDC-0310 (2), which possesses improved Nav selectivity and pharmacokinetic profile over 1.
Collapse
Affiliation(s)
- Brian S Safina
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Steven J McKerrall
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shaoyi Sun
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Chien-An Chen
- Chempartner, Building No. 5, 998 Halei Road, Zhangjiang Hi-Tech Park, Pudong New Area, Shanghai 201203, P.R. China
| | - Sultan Chowdhury
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Qi Jia
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Jun Li
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Alla Y Zenova
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Jean-Christophe Andrez
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Girish Bankar
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Philippe Bergeron
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jae H Chang
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Elaine Chang
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Jun Chen
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Richard Dean
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Shannon M Decker
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Antonio DiPasquale
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Thilo Focken
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Ivan Hemeon
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Kuldip Khakh
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Amy Kim
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Rainbow Kwan
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Andrea Lindgren
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Sophia Lin
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Jonathan Maher
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Janette Mezeyova
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Dinah Misner
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Karen Nelkenbrecher
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Jodie Pang
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Rebecca Reese
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shannon D Shields
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Luis Sojo
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Tao Sheng
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Henry Verschoof
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Matthew Waldbrook
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Michael S Wilson
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Zhiwei Xie
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Clint Young
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Tanja S Zabka
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - David H Hackos
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Daniel F Ortwine
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Andrew D White
- Chempartner, Building No. 5, 998 Halei Road, Zhangjiang Hi-Tech Park, Pudong New Area, Shanghai 201203, P.R. China
| | - J P Johnson
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - C Lee Robinette
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Christoph M Dehnhardt
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Charles J Cohen
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Daniel P Sutherlin
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
15
|
Sonvane S, Choudhari P, Bhusnuare O. In silico analysis of polyphenols and flavonoids for design of human Nav1.7 inhibitors. J Biomol Struct Dyn 2020; 39:4472-4479. [PMID: 32686994 DOI: 10.1080/07391102.2020.1777902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Neuropathic pain is commonly associated with lesion or disease of the somatosensory system and often reflected as indicator of impaired life. Although the central nervous system is main regulator of pain but for initiation and maintenance of the neuropathic pain is regulated by peripheral nervous system. Sodium channels particularly Nav1.7, Nav1.8, Nav 1.9 are key stake holders in the peripheral neuropathy, activation of these sodium channels might lead to genesis and propagation. Flavonoids and polyphenols showed promising effects in neuropathic pain. Here we are reporting In silico analysis of some selected flavonoids and polyphenols on sodium activated voltage channel 1.7 to explore the structural fragments required for binding. Results indicated Baicalin, Butrin, Dihydromonospermoside, Icariin, Isocoreopsin and Isosaponarin are showing promising docking score with sodium activated voltage channel 1.7 than other compounds. Structural modification of these promising leads keeping pharamcophoric requirement intact may yield potent Nav1.7 inhibitors for peripheral pain management.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sameep Sonvane
- Channabasweshwar Pharmacy College (Degree), Latur, India.,Department of Pharmaceutical Chemistry, Dayanand College of Pharmacy, Latur, India
| | - Prafulla Choudhari
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, India
| | - Omprakash Bhusnuare
- Channabasweshwar Pharmacy College (Degree), Latur, India.,Department of Pharmaceutical Chemistry, Dayanand College of Pharmacy, Latur, India
| |
Collapse
|
16
|
Merchant RR, Lang SB, Yu T, Zhao S, Qi Z, Suzuki T, Bao J. A General One-Pot Protocol for Hindered N-Alkyl Azaheterocycles from Tertiary Carboxylic Acids. Org Lett 2020; 22:4180-4184. [PMID: 32383385 DOI: 10.1021/acs.orglett.0c01254] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In this letter, we report a general one-pot strategy that utilizes three elementary steps (decarboxylative hydrazination, Boc deprotection, and heterocycle condensation) to regioselectively prepare hindered C(sp3) substituted pyrazoles and triazoles. The operational simplicity of this sequence and ubiquity of tertiary carboxylic acids allow rapid access to hindered N-alkyl azaheterocycles that will be useful to practitioners of medicinal chemistry and agro-chemistry.
Collapse
Affiliation(s)
- Rohan R Merchant
- Department of Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Simon B Lang
- Department of Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Tingting Yu
- IDSU, WuXi AppTec Co., Ltd., Shanghai 200131, China
| | | | - Zhiqi Qi
- IDSU, WuXi AppTec Co., Ltd., Shanghai 200131, China
| | - Takao Suzuki
- IDSU, WuXi AppTec Co., Ltd., Shanghai 200131, China
| | - Jianming Bao
- Department of Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| |
Collapse
|
17
|
Ramdas V, Talwar R, Kanoje V, Loriya RM, Banerjee M, Patil P, Joshi AA, Datrange L, Das AK, Walke DS, Kalhapure V, Khan T, Gote G, Dhayagude U, Deshpande S, Shaikh J, Chaure G, Pal RR, Parkale S, Suravase S, Bhoskar S, Gupta RV, Kalia A, Yeshodharan R, Azhar M, Daler J, Mali V, Sharma G, Kishore A, Vyawahare R, Agarwal G, Pareek H, Budhe S, Nayak A, Warude D, Gupta PK, Joshi P, Joshi S, Darekar S, Pandey D, Wagh A, Nigade PB, Mehta M, Patil V, Modi D, Pawar S, Verma M, Singh M, Das S, Gundu J, Nemmani K, Bock MG, Sharma S, Bakhle D, Kamboj RK, Palle VP. Discovery of Potent, Selective, and State-Dependent Na V1.7 Inhibitors with Robust Oral Efficacy in Pain Models: Structure-Activity Relationship and Optimization of Chroman and Indane Aryl Sulfonamides. J Med Chem 2020; 63:6107-6133. [PMID: 32368909 DOI: 10.1021/acs.jmedchem.0c00361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Voltage-gated sodium channel NaV1.7 is a genetically validated target for pain. Identification of NaV1.7 inhibitors with all of the desired properties to develop as an oral therapeutic for pain has been a major challenge. Herein, we report systematic structure-activity relationship (SAR) studies carried out to identify novel sulfonamide derivatives as potent, selective, and state-dependent NaV1.7 inhibitors for pain. Scaffold hopping from benzoxazine to chroman and indane bicyclic system followed by thiazole replacement on sulfonamide led to identification of lead molecules with significant improvement in solubility, selectivity over NaV1.5, and CYP2C9 inhibition. The lead molecules 13, 29, 32, 43, and 51 showed a favorable pharmacokinetics (PK) profile across different species and robust efficacy in veratridine and formalin-induced inflammatory pain models in mice. Compound 51 also showed significant effects on the CCI-induced neuropathic pain model. The profile of 51 indicated that it has the potential for further evaluation as a therapeutic for pain.
Collapse
Affiliation(s)
- Vidya Ramdas
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Rashmi Talwar
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Vijay Kanoje
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Rajesh M Loriya
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Moloy Banerjee
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Pradeep Patil
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Advait Arun Joshi
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Laxmikant Datrange
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Amit Kumar Das
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Deepak Sahebrao Walke
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Vaibhav Kalhapure
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Talha Khan
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Ganesh Gote
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Usha Dhayagude
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Shreyas Deshpande
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Javed Shaikh
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Ganesh Chaure
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Ravindra R Pal
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Santosh Parkale
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Sachin Suravase
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Smita Bhoskar
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Rajesh V Gupta
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Anil Kalia
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Rajesh Yeshodharan
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Mahammad Azhar
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Jagadeesh Daler
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Vinod Mali
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Geetika Sharma
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Amitesh Kishore
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Rupali Vyawahare
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Gautam Agarwal
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Himani Pareek
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Sagar Budhe
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Arun Nayak
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Dnyaneshwar Warude
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Praveen Kumar Gupta
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Parag Joshi
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Sneha Joshi
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Sagar Darekar
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Dilip Pandey
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Akshaya Wagh
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Prashant B Nigade
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Maneesh Mehta
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Vinod Patil
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Dipak Modi
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Shashikant Pawar
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Mahip Verma
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Minakshi Singh
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Sudipto Das
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Jayasagar Gundu
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Kumar Nemmani
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Mark G Bock
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Sharad Sharma
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Dhananjay Bakhle
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Rajender Kumar Kamboj
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| | - Venkata P Palle
- Novel Drug Discovery & Development, Lupin Ltd., Lupin Research Park, Survey No. 46 A/47 A, Village Nande, Taluka Mulshi, Pune 412115, India
| |
Collapse
|
18
|
Shinozuka T, Kobayashi H, Suzuki S, Tanaka K, Karanjule N, Hayashi N, Tsuda T, Tokumaru E, Inoue M, Ueda K, Kimoto H, Domon Y, Takahashi S, Kubota K, Yokoyama T, Shimizugawa A, Koishi R, Fujiwara C, Asano D, Sakakura T, Takasuna K, Abe Y, Watanabe T, Kitano Y. Discovery of DS-1971a, a Potent, Selective NaV1.7 Inhibitor. J Med Chem 2020; 63:10204-10220. [DOI: 10.1021/acs.jmedchem.0c00259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Tsuyoshi Shinozuka
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Hiroyuki Kobayashi
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Sayaka Suzuki
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Kyosuke Tanaka
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Narayan Karanjule
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Noriyuki Hayashi
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Toshifumi Tsuda
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Eri Tokumaru
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Masahiro Inoue
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Kiyono Ueda
- R&D Division, Daiichi Sankyo RD Novare Co., Ltd., 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Hiroko Kimoto
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yuki Domon
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Sakiko Takahashi
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Kazufumi Kubota
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Tomihisa Yokoyama
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Akiko Shimizugawa
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Ryuta Koishi
- R&D Division, Daiichi Sankyo RD Novare Co., Ltd., 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Chie Fujiwara
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Daigo Asano
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Tomoko Sakakura
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Kiyoshi Takasuna
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yasuyuki Abe
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Toshiyuki Watanabe
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yutaka Kitano
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| |
Collapse
|
19
|
Jo S, Bean BP. Lidocaine Binding Enhances Inhibition of Nav1.7 Channels by the Sulfonamide PF-05089771. Mol Pharmacol 2020; 97:377-383. [PMID: 32193331 DOI: 10.1124/mol.119.118380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/27/2020] [Indexed: 11/22/2022] Open
Abstract
PF-05089771 is an aryl sulfonamide Nav1.7 channel blocker that binds to the inactivated state of Nav1.7 channels with high affinity but binds only weakly to channels in the resting state. Such aryl sulfonamide Nav1.7 channel blockers bind to the extracellular surface of the S1-S4 voltage-sensor segment of homologous Domain 4, whose movement is associated with inactivation. This binding site is different from that of classic sodium channel inhibitors like lidocaine, which also bind with higher affinity to the inactivated state than the resting state but bind at a site within the pore of the channel. The common dependence on gating state with distinct binding sites raises the possibility that inhibition by aryl sulfonamides and by classic local anesthetics might show an interaction mediated by their mutual state dependence. We tested this possibility by examining the state-dependent inhibition by PF-05089771 and lidocaine of human Nav1.7 channels expressed in human embryonic kidney 293 cells. At -80 mV, where a small fraction of channels are in an inactivated state under drug-free conditions, inhibition by PF-05089771 was both enhanced and speeded in the presence of lidocaine. The results suggest that lidocaine binding to the channel enhances PF-05089771 inhibition by altering the equilibrium between resting states (with D4S4 in the inner position) and inactivated states (with D4S4 in the outer position). The gating state-mediated interaction between the compounds illustrates a principle applicable to many state-dependent agents. SIGNIFICANCE STATEMENT: The results show that lidocaine enhances the degree and rate of inhibition of Nav1.7 channels by the aryl sulfonamide compound PF-05089771, consistent with state-dependent binding by lidocaine increasing the fraction of channels presenting a high-affinity binding site for PF-05089771 and suggesting that combinations of agents targeted to the pore-region binding site of lidocaine and the external binding site of aryl sulfonamides may have synergistic actions.
Collapse
Affiliation(s)
- Sooyeon Jo
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
20
|
Wang JT, Zheng YM, Chen YT, Gu M, Gao ZB, Nan FJ. Discovery of aryl sulfonamide-selective Nav1.7 inhibitors with a highly hydrophobic ethanoanthracene core. Acta Pharmacol Sin 2020; 41:293-302. [PMID: 31316182 PMCID: PMC7471454 DOI: 10.1038/s41401-019-0267-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/30/2019] [Indexed: 01/19/2023] Open
Abstract
Nav1.7 channels are mainly distributed in the peripheral nervous system. Blockade of Nav1.7 channels with small-molecule inhibitors in humans might provide pain relief without affecting the central nervous system. Based on the facts that many reported Nav1.7-selective inhibitors contain aryl sulfonamide fragments, as well as a tricyclic antidepressant, maprotiline, has been found to inhibit Nav1.7 channels, we designed and synthesized a series of compounds with ethanoanthracene and aryl sulfonamide moieties. Their inhibitory activity on sodium channels were detected with electrophysiological techniques. We found that compound 10o potently inhibited Nav1.7 channels stably expressed in HEK293 cells (IC50 = 0.64 ± 0.30 nmol/L) and displayed a high Nav1.7/Nav1.5 selectivity. In mouse small-sized dorsal root ganglion neurons, compound 10o (10, 100 nmol/L) dose-dependently decreased the sodium currents and dramatically suppressed depolarizing current-elicited neuronal discharge. Preliminary in vivo experiments showed that compound 10o possessed good analgesic activity: in a mouse visceral pain model, administration of compound 10o (30−100 mg/kg, i.p.) effectively and dose-dependently suppressed acetic acid-induced writhing.
Collapse
|
21
|
Li ZM, Chen LX, Li H. Voltage-gated Sodium Channels and Blockers: An Overview and Where Will They Go? Curr Med Sci 2019; 39:863-873. [PMID: 31845216 DOI: 10.1007/s11596-019-2117-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 09/02/2019] [Indexed: 11/27/2022]
Abstract
Voltage-gated sodium (Nav) channels are critical players in the generation and propagation of action potentials by triggering membrane depolarization. Mutations in Nav channels are associated with a variety of channelopathies, which makes them relevant targets for pharmaceutical intervention. So far, the cryoelectron microscopic structure of the human Nav1.2, Nav1.4, and Nav1.7 has been reported, which sheds light on the molecular basis of functional mechanism of Nav channels and provides a path toward structure-based drug discovery. In this review, we focus on the recent advances in the structure, molecular mechanism and modulation of Nav channels, and state updated sodium channel blockers for the treatment of pathophysiology disorders and briefly discuss where the blockers may be developed in the future.
Collapse
Affiliation(s)
- Zhi-Mei Li
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li-Xia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Hua Li
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
22
|
Mulcahy JV, Pajouhesh H, Beckley JT, Delwig A, Bois JD, Hunter JC. Challenges and Opportunities for Therapeutics Targeting the Voltage-Gated Sodium Channel Isoform Na V1.7. J Med Chem 2019; 62:8695-8710. [PMID: 31012583 PMCID: PMC6786914 DOI: 10.1021/acs.jmedchem.8b01906] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Voltage-gated sodium ion channel subtype 1.7 (NaV1.7) is a high interest target for the discovery of non-opioid analgesics. Compelling evidence from human genetic data, particularly the finding that persons lacking functional NaV1.7 are insensitive to pain, has spurred considerable effort to develop selective inhibitors of this Na+ ion channel target as analgesic medicines. Recent clinical setbacks and disappointing performance of preclinical compounds in animal pain models, however, have led to skepticism around the potential of selective NaV1.7 inhibitors as human therapeutics. In this Perspective, we discuss the attributes and limitations of recently disclosed investigational drugs targeting NaV1.7 and review evidence that, by better understanding the requirements for selectivity and target engagement, the opportunity to deliver effective analgesic medicines targeting NaV1.7 endures.
Collapse
Affiliation(s)
- John V. Mulcahy
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - Hassan Pajouhesh
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - Jacob T. Beckley
- SiteOne Therapeutics, 351 Evergreen Drive, Suite B1, Bozeman, MT 59715
| | - Anton Delwig
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - J. Du Bois
- Stanford University, Lokey Chemistry and Biology, 337 Campus Drive, Stanford, CA 94305
| | - John C. Hunter
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| |
Collapse
|
23
|
Kong DJ, Wang Y, Wang HX, Wang MX, Wang J, Cheng MS. Molecular determinants for ligand binding at Nav1.4 and Nav1.7 channels: Experimental affinity results analyzed by molecular modeling. Comput Biol Chem 2019; 83:107132. [PMID: 31563636 DOI: 10.1016/j.compbiolchem.2019.107132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/11/2019] [Accepted: 09/18/2019] [Indexed: 12/16/2022]
Abstract
Here, we focused on exploring the selectivity mechanism against Nav1.7 over Nav1.4 due to different binding modes of two selected inhibitors. By the superposition of Nav1.7 and Nav1.4 proteins, we selected the most homologous chain of Nav1.7 with Nav1.4, defining the active site of Nav1.4-VSD4 based on the aryl sulfonamide binding site of Nav1.7-VSD4. Comparison of the conformations exhibited by Tyr1386 (Nav1.4) and Tyr1537 (Nav1.7) suggested that the steric hindrance caused by Tyr1386 owned primary influence on inhibition selectivity, which was further verified through molecular docking and MD simulation of two representative inhibitors. Our finding would be helpful for discovery of selective Nav1.7 inhibitors over Nav1.4.
Collapse
Affiliation(s)
- De-Jiang Kong
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Ying Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Han-Xun Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Ming-Xing Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jian Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Mao-Sheng Cheng
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| |
Collapse
|
24
|
Karaki F, Umemoto S, Ashizawa K, Oki T, Sato N, Ogino T, Ishibashi N, Someya R, Miyano K, Hirayama S, Uezono Y, Fujii H. A New Lead Identification Strategy: Screening an sp
3
‐rich and Lead‐like Compound Library Composed of 7‐Azanorbornane Derivatives. ChemMedChem 2019; 14:1840-1848. [DOI: 10.1002/cmdc.201900398] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/18/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Fumika Karaki
- Laboratory of Medicinal Chemistry School of Pharmacy Kitasato University 5-9-1, Shirokane, Minato-ku Tokyo 108-8641 Japan
- Medicinal Research Laboratories School of Pharmacy Kitasato University 5-9-1, Shirokane, Minato-ku Tokyo 108-8641 Japan
| | - Sho Umemoto
- Laboratory of Medicinal Chemistry School of Pharmacy Kitasato University 5-9-1, Shirokane, Minato-ku Tokyo 108-8641 Japan
| | - Karin Ashizawa
- Laboratory of Medicinal Chemistry School of Pharmacy Kitasato University 5-9-1, Shirokane, Minato-ku Tokyo 108-8641 Japan
- Division of Cancer Pathophysiology National Cancer Center Research Institute 5-1-1 Tsukiji, Chuo-ku Tokyo 104-0045 Japan
| | - Tomoya Oki
- Laboratory of Medicinal Chemistry School of Pharmacy Kitasato University 5-9-1, Shirokane, Minato-ku Tokyo 108-8641 Japan
| | - Noriko Sato
- Analytical Unit for Organic Chemistry Kitasato University 5-9-1, Shirokane, Minato-ku Tokyo 108-8641 Japan
| | - Takumi Ogino
- Laboratory of Medicinal Chemistry School of Pharmacy Kitasato University 5-9-1, Shirokane, Minato-ku Tokyo 108-8641 Japan
- Division of Cancer Pathophysiology National Cancer Center Research Institute 5-1-1 Tsukiji, Chuo-ku Tokyo 104-0045 Japan
| | - Naoto Ishibashi
- Laboratory of Medicinal Chemistry School of Pharmacy Kitasato University 5-9-1, Shirokane, Minato-ku Tokyo 108-8641 Japan
- Division of Cancer Pathophysiology National Cancer Center Research Institute 5-1-1 Tsukiji, Chuo-ku Tokyo 104-0045 Japan
| | - Ryoto Someya
- Laboratory of Medicinal Chemistry School of Pharmacy Kitasato University 5-9-1, Shirokane, Minato-ku Tokyo 108-8641 Japan
- Division of Cancer Pathophysiology National Cancer Center Research Institute 5-1-1 Tsukiji, Chuo-ku Tokyo 104-0045 Japan
| | - Kanako Miyano
- Division of Cancer Pathophysiology National Cancer Center Research Institute 5-1-1 Tsukiji, Chuo-ku Tokyo 104-0045 Japan
| | - Shigeto Hirayama
- Laboratory of Medicinal Chemistry School of Pharmacy Kitasato University 5-9-1, Shirokane, Minato-ku Tokyo 108-8641 Japan
- Medicinal Research Laboratories School of Pharmacy Kitasato University 5-9-1, Shirokane, Minato-ku Tokyo 108-8641 Japan
| | - Yasuhito Uezono
- Division of Cancer Pathophysiology National Cancer Center Research Institute 5-1-1 Tsukiji, Chuo-ku Tokyo 104-0045 Japan
- Division of Supportive Care Research Exploratory Oncology Research & Clinical Trial Center National Cancer Center 5-1-1 Tsukiji, Chuo-ku Tokyo 104-0045 Japan
- Innovation Center for Supportive, Palliative and Psychosocial Care National Cancer Center Hospital 5-1-1 Tsukiji, Chuo-ku Tokyo 104-0045 Japan
- Department of Comprehensive Oncology Graduate School of Biomedical Sciences Nagasaki University 1-12-4 Sakamoto Nagasaki 852-8523 Japan
| | - Hideaki Fujii
- Laboratory of Medicinal Chemistry School of Pharmacy Kitasato University 5-9-1, Shirokane, Minato-ku Tokyo 108-8641 Japan
- Medicinal Research Laboratories School of Pharmacy Kitasato University 5-9-1, Shirokane, Minato-ku Tokyo 108-8641 Japan
| |
Collapse
|
25
|
Xu L, Ding X, Wang T, Mou S, Sun H, Hou T. Voltage-gated sodium channels: structures, functions, and molecular modeling. Drug Discov Today 2019; 24:1389-1397. [PMID: 31129313 DOI: 10.1016/j.drudis.2019.05.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/02/2019] [Accepted: 05/17/2019] [Indexed: 10/26/2022]
Abstract
Voltage-gated sodium channels (VGSCs), formed by 24 transmembrane segments arranged into four domains, have a key role in the initiation and propagation of electrical signaling in excitable cells. VGSCs are involved in a variety of diseases, including epilepsy, cardiac arrhythmias, and neuropathic pain, and therefore have been regarded as appealing therapeutic targets for the development of anticonvulsant, antiarrhythmic, and local anesthetic drugs. In this review, we discuss recent advances in understanding the structures and biological functions of VGSCs. In addition, we systematically summarize eight pharmacologically distinct ligand-binding sites in VGSCs and representative isoform-selective VGSC modulators in clinical trials. Finally, we review studies on molecular modeling and computer-aided drug design (CADD) for VGSCs to help understanding of biological processes involving VGSCs.
Collapse
Affiliation(s)
- Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Xiaoqin Ding
- Beijing Institute of Pharmaceutical Chemistry, Beijing 102205, China
| | - Tianhu Wang
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Shanzhi Mou
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou 213001, China
| | - Huiyong Sun
- Department of Medicinal Chemistry, School of Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
26
|
McKerrall SJ, Nguyen T, Lai KW, Bergeron P, Deng L, DiPasquale A, Chang JH, Chen J, Chernov-Rogan T, Hackos DH, Maher J, Ortwine DF, Pang J, Payandeh J, Proctor WR, Shields SD, Vogt J, Ji P, Liu W, Ballini E, Schumann L, Tarozzo G, Bankar G, Chowdhury S, Hasan A, Johnson JP, Khakh K, Lin S, Cohen CJ, Dehnhardt CM, Safina BS, Sutherlin DP. Structure- and Ligand-Based Discovery of Chromane Arylsulfonamide Nav1.7 Inhibitors for the Treatment of Chronic Pain. J Med Chem 2019; 62:4091-4109. [DOI: 10.1021/acs.jmedchem.9b00141] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Steven J. McKerrall
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Teresa Nguyen
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Kwong Wah Lai
- WuXi AppTec Co., Ltd., 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, People’s Republic of China
| | - Philippe Bergeron
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Lunbin Deng
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Antonio DiPasquale
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jae H. Chang
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jun Chen
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Tania Chernov-Rogan
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - David H. Hackos
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jonathan Maher
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Daniel F. Ortwine
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jodie Pang
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jian Payandeh
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - William R. Proctor
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shannon D. Shields
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jennifer Vogt
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Pengfei Ji
- WuXi AppTec Co., Ltd., 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, People’s Republic of China
| | - Wenfeng Liu
- WuXi AppTec Co., Ltd., 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, People’s Republic of China
| | | | | | | | - Girish Bankar
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Sultan Chowdhury
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Abid Hasan
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - J. P. Johnson
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Kuldip Khakh
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Sophia Lin
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Charles J. Cohen
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Christoph M. Dehnhardt
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Brian S. Safina
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Daniel P. Sutherlin
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|