1
|
Al-Aqtash R, Collier DM. Ionotropic purinergic receptor 7 (P2X7) channel structure and pharmacology provides insight regarding non-nucleotide agonism. Channels (Austin) 2024; 18:2355150. [PMID: 38762911 PMCID: PMC11110710 DOI: 10.1080/19336950.2024.2355150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/10/2024] [Indexed: 05/21/2024] Open
Abstract
P2X7 is a member of the Ionotropic Purinergic Receptor (P2X) family. The P2X family of receptors is composed of seven (P2X1-7), ligand-gated, nonselective cation channels. Changes in P2X expression have been reported in multiple disease models. P2Xs have large complex extracellular domains that function as receptors for a variety of ligands, including endogenous and synthetic agonists and antagonists. ATP is the canonical agonist. ATP affinity ranges from nanomolar to micromolar for most P2XRs, but P2X7 has uniquely poor ATP affinity. In many physiological settings, it may be difficult to achieve the millimolar extracellular ATP concentrations needed for P2X7 channel activation; however, channel function is implicated in pain sensation, immune cell function, cardiovascular disease, cancer, and osteoporosis. Multiple high-resolution P2X7 structures have been solved in apo-, ATP-, and antagonist-bound states. P2X7 structural data reveal distinct allosteric and orthosteric antagonist-binding sites. Both allosteric and orthosteric P2X7 antagonists are well documented to inhibit ATP-evoked channel current. However, a growing body of evidence supports P2X7 activation by non-nucleotide agonists, including extracellular histone proteins and human cathelicidin-derived peptides (LL-37). Interestingly, P2X7 non-nucleotide agonism is not inhibited by allosteric antagonists, but is inhibited by orthosteric antagonists. Herein, we review P2X7 function with a focus on the efficacy of available pharmacology on P2X7 channel current activation by non-nucleotide agonists in effort to understand agonist/antagonist efficacy, and consider the impact of these data on the current understanding of P2X7 in physiology and disease given these limitations of P2X7-selective antagonists and incomplete knockout mouse models.
Collapse
Affiliation(s)
- Rua’a Al-Aqtash
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Daniel M. Collier
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
2
|
Liu X, Li Y, Huang L, Kuang Y, Wu X, Ma X, Zhao B, Lan J. Unlocking the therapeutic potential of P2X7 receptor: a comprehensive review of its role in neurodegenerative disorders. Front Pharmacol 2024; 15:1450704. [PMID: 39139642 PMCID: PMC11319138 DOI: 10.3389/fphar.2024.1450704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
The P2X7 receptor (P2X7R), an ATP-gated ion channel, has emerged as a crucial player in neuroinflammation and a promising therapeutic target for neurodegenerative disorders. This review explores the current understanding of P2X7R's structure, activation, and physiological roles, focusing on its expression and function in microglial cells. The article examines the receptor's involvement in calcium signaling, microglial activation, and polarization, as well as its role in the pathogenesis of Alzheimer's disease, Parkinson's disease, multiple sclerosis, and amyotrophic lateral sclerosis. The review highlights the complex nature of P2X7R signaling, discussing its potential neuroprotective and neurotoxic effects depending on the disease stage and context. It also addresses the development of P2X7R antagonists and their progress in clinical trials, identifying key research gaps and future perspectives for P2X7R-targeted therapy development. By providing a comprehensive overview of the current state of knowledge and future directions, this review serves as a valuable resource for researchers and clinicians interested in exploring the therapeutic potential of targeting P2X7R for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Xiaoming Liu
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Yiwen Li
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Liting Huang
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Yingyan Kuang
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Xiaoxiong Wu
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Xiangqiong Ma
- Henan Hospital of Integrated Chinese and Western Medicine, Zhengzhou, China
| | - Beibei Zhao
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Jiao Lan
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| |
Collapse
|
3
|
Zhang R, Su K, Yang L, Tang M, Zhao M, Ye N, Cai X, Jiang X, Li N, Peng J, Zhang X, Wang B, Wu W, Ma L, Ye H. Design, Synthesis, and Biological Evaluation of Novel P2X7 Receptor Antagonists for the Treatment of Septic Acute Kidney Injury. J Med Chem 2023; 66:11365-11389. [PMID: 37582195 DOI: 10.1021/acs.jmedchem.3c00837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
Sepsis-associated acute kidney injury (AKI) is a serious clinical problem, without effective drugs. Abnormal activation of the purinergic P2X7 receptor (P2X7R) in septic kidneys makes its antagonist a promising therapeutic approach. Herein, a series of novel P2X7R antagonists were designed, synthesized, and structurally optimized. Based on in vitro potency in human/mouse P2X7R using HEK293 cells, hepatic microsomal stability, and pharmacokinetic and preliminary in vivo assessments, compound 14a was identified by respective human and mouse P2X7R IC50 values of 64.7 and 10.1 nM, together with favorable pharmacokinetic properties. Importantly, 14a dose-dependently alleviated kidney dysfunction and pathological injury in both lipopolysaccharide (LPS)- and cecal ligation/perforation (CLP)-induced septic AKI mice with a good safety profile. Mechanistically, 14a could suppress NLRP3 inflammasome activation to inhibit the expression of cleaved caspase-1, gasdermin D, IL-1β, and IL-18 in the injured kidneys of septic mice. Collectively, these results highlighted that P2X7R antagonist 14a exerted a therapeutic potential against septic AKI.
Collapse
Affiliation(s)
- Ruijia Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kaiyue Su
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Letian Yang
- Division of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Minghai Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Min Zhao
- Laboratory of Metabolomics and Drug-induced Liver Injury, Department of Gastroenterology & Hepatology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Neng Ye
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoying Cai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xueqin Jiang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Peng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinlu Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Wang
- Division of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenshuang Wu
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liang Ma
- Division of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haoyu Ye
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
4
|
Lukhey MS, Shende P. Advancement in wound healing treatment using functional nanocarriers. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2099393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Mihir S. Lukhey
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS, Mumbai, India
| | - Pravin Shende
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS, Mumbai, India
| |
Collapse
|
5
|
Rawish E, Langer HF. Platelets and the Role of P2X Receptors in Nociception, Pain, Neuronal Toxicity and Thromboinflammation. Int J Mol Sci 2022; 23:6585. [PMID: 35743029 PMCID: PMC9224425 DOI: 10.3390/ijms23126585] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 12/24/2022] Open
Abstract
P2X receptors belong to a family of cation channel proteins, which respond to extracellular adenosine 5'-triphosphate (ATP). These receptors have gained increasing attention in basic and translational research, as they are central to a variety of important pathophysiological processes such as the modulation of cardiovascular physiology, mediation of nociception, platelet and macrophage activation, or neuronal-glial integration. While P2X1 receptor activation is long known to drive platelet aggregation, P2X7 receptor antagonists have recently been reported to inhibit platelet activation. Considering the role of both P2X receptors and platelet-mediated inflammation in neuronal diseases such as multiple sclerosis, Alzheimer's disease, Parkinson's disease, and stroke, targeting purinergic receptors may provide a valuable novel therapeutic approach in these diseases. Therefore, the present review illuminates the role of platelets and purinergic signaling in these neurological conditions to evaluate potential translational implications.
Collapse
Affiliation(s)
- Elias Rawish
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, 23538 Lübeck, Germany;
- University Hospital Schleswig-Holstein, Department of Cardiology, University Heart Center Lübeck, 23538 Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, 23562 Lübeck, Germany
| | - Harald F. Langer
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, 23538 Lübeck, Germany;
- University Hospital Schleswig-Holstein, Department of Cardiology, University Heart Center Lübeck, 23538 Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, 23562 Lübeck, Germany
| |
Collapse
|
6
|
Calzaferri F, Narros-Fernández P, de Pascual R, de Diego AMG, Nicke A, Egea J, García AG, de Los Ríos C. Synthesis and Pharmacological Evaluation of Novel Non-nucleotide Purine Derivatives as P2X7 Antagonists for the Treatment of Neuroinflammation. J Med Chem 2021; 64:2272-2290. [PMID: 33560845 DOI: 10.1021/acs.jmedchem.0c02145] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ATP-gated P2X7 purinergic receptor (P2X7) is involved in the pathogenesis of many neurodegenerative diseases (NDDs). Several P2X7 antagonists have been developed, though none of them reached clinical trials for this indication. In this work, we designed and synthesized novel blood-brain barrier (BBB)-permeable derivatives as potential P2X7 antagonists. They comprise purine or xanthine cores linked to an aryl group through different short spacers. Compounds were tested in YO-PRO-1 uptake assays and intracellular calcium dynamics in a human P2X7-expressing HEK293 cell line, two-electrode voltage-clamp recordings in Xenopus laevis oocytes, and in interleukin 1β release assays in mouse peritoneal macrophages. BBB permeability was assessed by parallel artificial membrane permeability assays and P-glycoprotein ATPase activity. Dichloroarylpurinylethanones featured a certain P2X7 blockade, being compound 6 (2-(6-chloro-9H-purin-9-yl)-1-(2,4-dichlorophenyl)ethan-1-one), named ITH15004, the most potent, selective, and BBB-permeable antagonist. Compound 6 can be considered as a first non-nucleotide purine hit for future drug optimizations.
Collapse
Affiliation(s)
- Francesco Calzaferri
- Instituto-Fundación Teófilo Hernando and Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Paloma Narros-Fernández
- Instituto-Fundación Teófilo Hernando and Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo 4, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, C/ Diego de León, 62-1a Planta, 28006 Madrid, Spain
| | - Ricardo de Pascual
- Instituto-Fundación Teófilo Hernando and Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Antonio M G de Diego
- Instituto-Fundación Teófilo Hernando and Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Annette Nicke
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| | - Javier Egea
- Instituto-Fundación Teófilo Hernando and Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo 4, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, C/ Diego de León, 62-1a Planta, 28006 Madrid, Spain
| | - Antonio G García
- Instituto-Fundación Teófilo Hernando and Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo 4, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, C/ Diego de León, 62-1a Planta, 28006 Madrid, Spain
| | - Cristóbal de Los Ríos
- Instituto-Fundación Teófilo Hernando and Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo 4, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, C/ Diego de León, 62-1a Planta, 28006 Madrid, Spain
| |
Collapse
|
7
|
Suresh P, Phasuk S, Liu IY. Modulation of microglia activation and Alzheimer's disease: CX3 chemokine ligand 1/CX3CR and P2X 7R signaling. Tzu Chi Med J 2021; 33:1-6. [PMID: 33505871 PMCID: PMC7821819 DOI: 10.4103/tcmj.tcmj_144_20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/05/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive deficits. Two hallmarks of AD that cause chronic inflammation and lead to neuronal dysfunction and damage are tau tangles and amyloid plaques. Microglial cells, the primary immune cells of the central nervous system, maintain a homeostatic active/inactive state via a bidirectional, dynamic communication with neurons. Several studies have revealed that dysregulated microglial activation leads to AD pathology. Therefore, we reviewed the relationship between AD and two important signaling complexes, CX3 chemokine ligand 1 (CX3CL1)/CX3CR1 and ATP/P2X7R, that play critical roles in the regulation of microglial activation. CX3CL1/CX3CR1 is one important signaling which controls the microglia function. Altering this pathway can have opposite effects on amyloid and tau pathology in AD. Another important molecule is P2X7R which involves in the activation of microglia. Over activation of P2X7R is evident in AD pathogenesis. In this review, we discuss influence of the two signaling pathways at different stages of AD pathology as well as the drug candidates that can modulate CX3CL1/CX3CR1 and ATP/P2X7R.
Collapse
Affiliation(s)
- Pavithra Suresh
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Sarayut Phasuk
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Ingrid Y Liu
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
8
|
P2X7 Receptors Amplify CNS Damage in Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21175996. [PMID: 32825423 PMCID: PMC7504621 DOI: 10.3390/ijms21175996] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
ATP is a (co)transmitter and signaling molecule in the CNS. It acts at a multitude of ligand-gated cationic channels termed P2X to induce rapid depolarization of the cell membrane. Within this receptor-channel family, the P2X7 receptor (R) allows the transmembrane fluxes of Na+, Ca2+, and K+, but also allows the slow permeation of larger organic molecules. This is supposed to cause necrosis by excessive Ca2+ influx, as well as depletion of intracellular ions and metabolites. Cell death may also occur by apoptosis due to the activation of the caspase enzymatic cascade. Because P2X7Rs are localized in the CNS preferentially on microglia, but also at a lower density on neuroglia (astrocytes, oligodendrocytes) the stimulation of this receptor leads to the release of neurodegeneration-inducing bioactive molecules such as pro-inflammatory cytokines, chemokines, proteases, reactive oxygen and nitrogen molecules, and the excitotoxic glutamate/ATP. Various neurodegenerative reactions of the brain/spinal cord following acute harmful events (mechanical CNS damage, ischemia, status epilepticus) or chronic neurodegenerative diseases (neuropathic pain, Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, amyotrophic lateral sclerosis) lead to a massive release of ATP via the leaky plasma membrane of neural tissue. This causes cellular damage superimposed on the original consequences of neurodegeneration. Hence, blood-brain-barrier permeable pharmacological antagonists of P2X7Rs with excellent bioavailability are possible therapeutic agents for these diseases. The aim of this review article is to summarize our present state of knowledge on the involvement of P2X7R-mediated events in neurodegenerative illnesses endangering especially the life quality and duration of the aged human population.
Collapse
|
9
|
Andrejew R, Oliveira-Giacomelli Á, Ribeiro DE, Glaser T, Arnaud-Sampaio VF, Lameu C, Ulrich H. The P2X7 Receptor: Central Hub of Brain Diseases. Front Mol Neurosci 2020; 13:124. [PMID: 32848594 PMCID: PMC7413029 DOI: 10.3389/fnmol.2020.00124] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/17/2020] [Indexed: 12/27/2022] Open
Abstract
The P2X7 receptor is a cation channel activated by high concentrations of adenosine triphosphate (ATP). Upon long-term activation, it complexes with membrane proteins forming a wide pore that leads to cell death and increased release of ATP into the extracellular milieu. The P2X7 receptor is widely expressed in the CNS, such as frontal cortex, hippocampus, amygdala and striatum, regions involved in neurodegenerative diseases and psychiatric disorders. Despite P2X7 receptor functions in glial cells have been extensively studied, the existence and roles of this receptor in neurons are still controversially discussed. Regardless, P2X7 receptors mediate several processes observed in neuropsychiatric disorders and brain tumors, such as activation of neuroinflammatory response, stimulation of glutamate release and neuroplasticity impairment. Moreover, P2X7 receptor gene polymorphisms have been associated to depression, and isoforms of P2X7 receptors are implicated in neuropsychiatric diseases. In view of that, the P2X7 receptor has been proposed to be a potential target for therapeutic intervention in brain diseases. This review discusses the molecular mechanisms underlying P2X7 receptor-mediated signaling in neurodegenerative diseases, psychiatric disorders, and brain tumors. In addition, it highlights the recent advances in the development of P2X7 receptor antagonists that are able of penetrating the central nervous system.
Collapse
Affiliation(s)
- Roberta Andrejew
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | - Deidiane Elisa Ribeiro
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Talita Glaser
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | - Claudiana Lameu
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
Calzaferri F, Ruiz-Ruiz C, de Diego AMG, de Pascual R, Méndez-López I, Cano-Abad MF, Maneu V, de Los Ríos C, Gandía L, García AG. The purinergic P2X7 receptor as a potential drug target to combat neuroinflammation in neurodegenerative diseases. Med Res Rev 2020; 40:2427-2465. [PMID: 32677086 DOI: 10.1002/med.21710] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 06/22/2020] [Accepted: 06/27/2020] [Indexed: 12/25/2022]
Abstract
Neurodegenerative diseases (NDDs) represent a huge social burden, particularly in Alzheimer's disease (AD) in which all proposed treatments investigated in murine models have failed during clinical trials (CTs). Thus, novel therapeutic strategies remain crucial. Neuroinflammation is a common pathogenic feature of NDDs. As purinergic P2X7 receptors (P2X7Rs) are gatekeepers of inflammation, they could be developed as drug targets for NDDs. Herein, we review this challenging hypothesis and comment on the numerous studies that have investigated P2X7Rs, emphasizing their molecular structure and functions, as well as their role in inflammation. Then, we elaborate on research undertaken in the field of medicinal chemistry to determine potential P2X7R antagonists. Subsequently, we review the state of neuroinflammation and P2X7R expression in the brain, in animal models and patients suffering from AD, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, multiple sclerosis, and retinal degeneration. Next, we summarize the in vivo studies testing the hypothesis that by mitigating neuroinflammation, P2X7R blockers afford neuroprotection, increasing neuroplasticity and neuronal repair in animal models of NDDs. Finally, we reviewed previous and ongoing CTs investigating compounds directed toward targets associated with NDDs; we propose that CTs with P2X7R antagonists should be initiated. Despite the high expectations for putative P2X7Rs antagonists in various central nervous system diseases, the field is moving forward at a relatively slow pace, presumably due to the complexity of P2X7Rs. A better pharmacological approach to combat NDDs would be a dual strategy, combining P2X7R antagonism with drugs targeting a selective pathway in a given NDD.
Collapse
Affiliation(s)
- Francesco Calzaferri
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Cristina Ruiz-Ruiz
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio M G de Diego
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ricardo de Pascual
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Iago Méndez-López
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - María F Cano-Abad
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Victoria Maneu
- Departamento de Óptica, Farmacología y Anatomía, Universidad de Alicante, San Vicente del Raspeig, Spain
| | - Cristóbal de Los Ríos
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Luis Gandía
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio G García
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
11
|
Epplin MP, Mohan A, Harris LD, Zhu Z, Strong KL, Bacsa J, Le P, Menaldino DS, Traynelis SF, Liotta DC. Discovery of Dihydropyrrolo[1,2- a]pyrazin-3(4 H)-one-Based Second-Generation GluN2C- and GluN2D-Selective Positive Allosteric Modulators (PAMs) of the N-Methyl-d-Aspartate (NMDA) Receptor. J Med Chem 2020; 63:7569-7600. [PMID: 32538088 DOI: 10.1021/acs.jmedchem.9b01733] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The N-methyl-d-aspartate receptor (NMDAR) is an ion channel that mediates the slow, Ca2+-permeable component of glutamatergic synaptic transmission in the central nervous system (CNS). NMDARs are known to play a significant role in basic neurological functions, and their dysfunction has been implicated in several CNS disorders. Herein, we report the discovery of second-generation GluN2C/D-selective NMDAR-positive allosteric modulators (PAMs) with a dihydropyrrolo[1,2-a]pyrazin-3(4H)-one core. The prototype, R-(+)-EU-1180-453, exhibits log unit improvements in the concentration needed to double receptor response, lipophilic efficiency, and aqueous solubility, and lowers cLogP by one log unit compared to the first-generation prototype CIQ. Additionally, R-(+)-EU-1180-453 was found to increase glutamate potency 2-fold, increase the response to maximally effective concentration of agonist 4-fold, and the racemate is brain-penetrant. These compounds are useful second-generation in vitro tools and a promising step toward in vivo tools for the study of positive modulation of GluN2C- and GluN2D-containing NMDA receptors.
Collapse
Affiliation(s)
- Matthew P Epplin
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Ayush Mohan
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Lynnea D Harris
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Zongjian Zhu
- Department of Pharmacology and Chemical Biology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Katie L Strong
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - John Bacsa
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Phuong Le
- Department of Pharmacology and Chemical Biology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - David S Menaldino
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Dennis C Liotta
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| |
Collapse
|
12
|
Dos Santos EG, Faria RX, Rodrigues CR, Bello ML. Molecular dynamic simulations of full-length human purinergic receptor subtype P2X7 bonded to potent inhibitors. Eur J Pharm Sci 2020; 152:105454. [PMID: 32629018 DOI: 10.1016/j.ejps.2020.105454] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/12/2020] [Accepted: 07/02/2020] [Indexed: 01/11/2023]
Abstract
Among the members of purinergic receptors, the family P2X of ionotropic proteins has the ion channel subtype P2X7 that show in studies to be an important molecular target for new drugs. The activity of human P2X7 receptor (hP2X7r) in the body, due to its pro-inflammatory function, can trigger physiological disorders related to chronic inflammatory processes, leading to neural degeneration, neuropathic pain and chronic pain. Recently, two series of promising new inhibitors of the hP2X7r ion channel have been reported. One series consisted of naphthoquinone derivatives and the other composed of triazole derivatives. The main objective of this study was to understand the binding mode differences between the hit compounds of each series and compare them to the native ligand ATP. The hP2X7r ion channel and membrane lipid models were prepared in order to allow study the appropriate protein molecular dynamics. Molecular modeling and molecular dynamics simulation approaches were applied in order to obtain atomistic and molecular details that are involved in intermolecular interactions. Both compounds AN-04 and 9d seem to have affinity to binding in the hP2X7r pore area according to molecular dynamics simulations results. The naphthoquinone derivative AN-04 demonstrated a binding free energy 7.68 fold larger than triazole derivative 9d and 3.8 fold lower than native ligand ATP. These results indicate that compound AN-04 might be a promising lead compound for the development of a novel selective hP2X7r inhibitor.
Collapse
Affiliation(s)
- Eldio G Dos Santos
- Laboratório de Planejamento Farmacêutico e Simulação Computacional, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robson X Faria
- Laboratório de Toxoplasmose e outras Protozooses, Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Carlos R Rodrigues
- ModMolQSAR, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Murilo L Bello
- Laboratório de Planejamento Farmacêutico e Simulação Computacional, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
13
|
Homerin G, Jawhara S, Dezitter X, Baudelet D, Dufrénoy P, Rigo B, Millet R, Furman C, Ragé G, Lipka E, Farce A, Renault N, Sendid B, Charlet R, Leroy J, Phanithavong M, Richeval C, Wiart JF, Allorge D, Adriouch S, Vouret-Craviari V, Ghinet A. Pyroglutamide-Based P2X7 Receptor Antagonists Targeting Inflammatory Bowel Disease. J Med Chem 2019; 63:2074-2094. [PMID: 31525963 DOI: 10.1021/acs.jmedchem.9b00584] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This report deals with the design, the synthesis, and the pharmacological evaluation of pyroglutamide-based P2X7 antagonists. A dozen were shown to possess improved properties, among which inhibition of YO-PRO-1/TO-PRO-3 uptake and IL1β release upon BzATP activation of the receptor and dampening signs of DSS-induced colitis on mice, in comparison with reference antagonist GSK1370319A. Docking study and biological evaluation of synthesized compounds has highlighted new SAR, and low toxicity profiles of pyroglutamides herein described are clues for the finding of a usable h-P2X7 antagonist drug. Such a drug would raise the hope for a cure to many P2X7-dependent pathologies, including inflammatory, neurological, and immune diseases.
Collapse
Affiliation(s)
- Germain Homerin
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Yncréa Hauts-de-France, UCLille, Laboratoire de Pharmacochimie, Hautes Etudes d'Ingénieur (HEI), 13 rue de Toul, F-59046 Lille, France
| | - Samir Jawhara
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France
| | - Xavier Dezitter
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Institut de Chimie Pharmaceutique Albert Lespagnol, IFR114, 3 rue du Pr Laguesse, F-59006 Lille, France
| | - Davy Baudelet
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Yncréa Hauts-de-France, UCLille, Laboratoire de Pharmacochimie, Hautes Etudes d'Ingénieur (HEI), 13 rue de Toul, F-59046 Lille, France
| | - Pierrick Dufrénoy
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Yncréa Hauts-de-France, UCLille, Laboratoire de Pharmacochimie, Hautes Etudes d'Ingénieur (HEI), 13 rue de Toul, F-59046 Lille, France
| | - Benoît Rigo
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Yncréa Hauts-de-France, UCLille, Laboratoire de Pharmacochimie, Hautes Etudes d'Ingénieur (HEI), 13 rue de Toul, F-59046 Lille, France
| | - Régis Millet
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Institut de Chimie Pharmaceutique Albert Lespagnol, IFR114, 3 rue du Pr Laguesse, F-59006 Lille, France
| | - Christophe Furman
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Institut de Chimie Pharmaceutique Albert Lespagnol, IFR114, 3 rue du Pr Laguesse, F-59006 Lille, France
| | - Guillaume Ragé
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Institut de Chimie Pharmaceutique Albert Lespagnol, IFR114, 3 rue du Pr Laguesse, F-59006 Lille, France
| | - Emmanuelle Lipka
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Laboratoire de Chimie Analytique, Faculté des Sciences Pharmaceutiques et Biologiques de Lille, F-59006 Lille Cedex, France
| | - Amaury Farce
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Institut de Chimie Pharmaceutique Albert Lespagnol, IFR114, 3 rue du Pr Laguesse, F-59006 Lille, France
| | - Nicolas Renault
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Institut de Chimie Pharmaceutique Albert Lespagnol, IFR114, 3 rue du Pr Laguesse, F-59006 Lille, France
| | - Boualem Sendid
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France
| | - Rogatien Charlet
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France
| | - Jordan Leroy
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France
| | - Mélodie Phanithavong
- Laboratoire de Toxicologie & Génopathies, CHRU de Lille, Centre de Biologie Pathologie, Blvd du Pr. J. Leclercq, CS 70001, F-59037 Lille, France
| | - Camille Richeval
- Laboratoire de Toxicologie & Génopathies, CHRU de Lille, Centre de Biologie Pathologie, Blvd du Pr. J. Leclercq, CS 70001, F-59037 Lille, France
| | - Jean-François Wiart
- Laboratoire de Toxicologie & Génopathies, CHRU de Lille, Centre de Biologie Pathologie, Blvd du Pr. J. Leclercq, CS 70001, F-59037 Lille, France
| | - Delphine Allorge
- Laboratoire de Toxicologie & Génopathies, CHRU de Lille, Centre de Biologie Pathologie, Blvd du Pr. J. Leclercq, CS 70001, F-59037 Lille, France
| | - Sahil Adriouch
- INSERM U905, F-76183 Rouen, France.,Institute for Research and Innovation in Biomedicine, Normandie University, F-76183 Rouen, France
| | - Valérie Vouret-Craviari
- Institute for Research on Cancer and Aging (IRCAN), F-06100 Nice, France.,University of Nice Cote d'Azur (UCA), F-06100 Nice, France
| | - Alina Ghinet
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Yncréa Hauts-de-France, UCLille, Laboratoire de Pharmacochimie, Hautes Etudes d'Ingénieur (HEI), 13 rue de Toul, F-59046 Lille, France.,Faculty of Chemistry, "Al. I. Cuza" University of Iasi, Blvd Carol I, nr. 11, 700506 Iasi, Romania
| |
Collapse
|
14
|
Rifampicin and Its Derivative Rifampicin Quinone Reduce Microglial Inflammatory Responses and Neurodegeneration Induced In Vitro by α-Synuclein Fibrillary Aggregates. Cells 2019; 8:cells8080776. [PMID: 31349736 PMCID: PMC6721546 DOI: 10.3390/cells8080776] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/12/2019] [Accepted: 07/18/2019] [Indexed: 12/22/2022] Open
Abstract
: Aggregated forms of the synaptic protein α-synuclein (αS) have been proposed to operate as a molecular trigger for microglial inflammatory processes and neurodegeneration in Parkinson´s disease. Here, we used brain microglial cell cultures activated by fibrillary forms of recombinant human αS to assess the anti-inflammatory and neuroprotective activities of the antibiotic rifampicin (Rif) and its autoxidation product rifampicin quinone (RifQ). Pretreatments with Rif and RifQ reduced the secretion of prototypical inflammatory cytokines (TNF-, IL-6) and the burst of oxidative stress in microglial cells activated with αS fibrillary aggregates. Note, however, that RifQ was constantly more efficacious than its parent compound in reducing microglial activation. We also established that the suppressive effects of Rif and RifQ on cytokine release was probably due to inhibition of both PI3K- and non-PI3K-dependent signaling events. The control of oxidative stress appeared, however, essentially dependent on PI3K inhibition. Of interest, we also showed that RifQ was more efficient than Rif in protecting neuronal cells from toxic factors secreted by microglia activated by αS fibrils. Overall, data with RifQ are promising enough to justify further studies to confirm the potential of this compound as an anti-parkinsionian drug.
Collapse
|
15
|
Inhibitors of NF-κB and P2X7/NLRP3/Caspase 1 pathway in microglia: Novel therapeutic opportunities in neuroinflammation induced early-stage Alzheimer’s disease. J Neuroimmunol 2019; 326:62-74. [DOI: 10.1016/j.jneuroim.2018.11.010] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/16/2018] [Accepted: 11/18/2018] [Indexed: 12/21/2022]
|
16
|
Dos-Santos-Pereira M, Acuña L, Hamadat S, Rocca J, González-Lizárraga F, Chehín R, Sepulveda-Diaz J, Del-Bel E, Raisman-Vozari R, Michel PP. Microglial glutamate release evoked by α-synuclein aggregates is prevented by dopamine. Glia 2018; 66:2353-2365. [PMID: 30394585 DOI: 10.1002/glia.23472] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/17/2018] [Accepted: 05/22/2018] [Indexed: 01/11/2023]
Abstract
When activated, microglial cells have the potential not only to secrete typical proinflammatory mediators but also to release the neurotransmitter glutamate in amounts that may promote excitotoxicity. Here, we wished to determine the potential of the Parkinson's disease (PD) protein α-Synuclein (αS) to stimulate glutamate release using cultures of purified microglial cells. We established that glutamate release was robustly increased when microglial cultures were treated with fibrillary aggregates of αS but not with the native monomeric protein. Promotion of microglial glutamate release by αS aggregates (αSa) required concomitant engagement of TLR2 and P2X7 receptors. Downstream to cell surface receptors, the release process was mediated by activation of a signaling cascade sequentially involving phosphoinositide 3-kinase (PI3K) and NADPH oxidase, a superoxide-producing enzyme. Inhibition of the Xc- antiporter, a plasma membrane exchange system that imports extracellular l-cystine and exports intracellular glutamate, prevented the release of glutamate induced by αSa, indicating that system Xc- was the final effector element in the release process downstream to NADPH oxidase activation. Of interest, the stimulation of glutamate release by αSa was abrogated by dopamine through an antioxidant effect requiring D1 dopamine receptor activation and PI3K inhibition. Altogether, present data suggest that the activation of microglial cells by αSa may possibly result in a toxic build-up of extracellular glutamate contributing to excitotoxic stress in PD. The deficit in dopamine that characterizes this disorder may further aggravate this process in a vicious circle mechanism.
Collapse
Affiliation(s)
- Mauricio Dos-Santos-Pereira
- Institut du Cerveau et de la Moelle épinière (ICM), Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Paris, F-75013, France.,Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil
| | - Leonardo Acuña
- Institut du Cerveau et de la Moelle épinière (ICM), Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Paris, F-75013, France.,Instituto de Patología Experimental (CONICET-UNSa), Salta, Argentina
| | - Sabah Hamadat
- Institut du Cerveau et de la Moelle épinière (ICM), Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Paris, F-75013, France
| | - Jeremy Rocca
- Institut du Cerveau et de la Moelle épinière (ICM), Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Paris, F-75013, France
| | - Florencia González-Lizárraga
- Institut du Cerveau et de la Moelle épinière (ICM), Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Paris, F-75013, France.,Instituto de Medicina Molecular y Celular Aplicada (IMMCA) CONICET/UNT and SIPROSA, Tucumán, Argentina
| | - Rosana Chehín
- Instituto de Medicina Molecular y Celular Aplicada (IMMCA) CONICET/UNT and SIPROSA, Tucumán, Argentina
| | - Julia Sepulveda-Diaz
- Institut du Cerveau et de la Moelle épinière (ICM), Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Paris, F-75013, France
| | - Elaine Del-Bel
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil
| | - Rita Raisman-Vozari
- Institut du Cerveau et de la Moelle épinière (ICM), Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Paris, F-75013, France
| | - Patrick P Michel
- Institut du Cerveau et de la Moelle épinière (ICM), Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Paris, F-75013, France
| |
Collapse
|
17
|
Janssen B, Vugts DJ, Wilkinson SM, Ory D, Chalon S, Hoozemans JJM, Schuit RC, Beaino W, Kooijman EJM, van den Hoek J, Chishty M, Doméné A, Van der Perren A, Villa A, Maggi A, Molenaar GT, Funke U, Shevchenko RV, Baekelandt V, Bormans G, Lammertsma AA, Kassiou M, Windhorst AD. Identification of the allosteric P2X 7 receptor antagonist [ 11C]SMW139 as a PET tracer of microglial activation. Sci Rep 2018; 8:6580. [PMID: 29700413 PMCID: PMC5920098 DOI: 10.1038/s41598-018-24814-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/15/2018] [Indexed: 02/07/2023] Open
Abstract
The P2X7 receptor plays a significant role in microglial activation, and as a potential drug target, the P2X7 receptor is also an interesting target in positron emission tomography. The current study aimed at the development and evaluation of a potent tracer targeting the P2X7 receptor, to which end four adamantanyl benzamide analogues with high affinity for the human P2X7 receptor were labelled with carbon-11. All four analogues could be obtained in excellent radiochemical yield and high radiochemical purity and molar activity, and all analogues entered the rat brain. [11C]SMW139 showed the highest metabolic stability in rat plasma, and showed high binding to the hP2X7 receptor in vivo in a hP2X7 receptor overexpressing rat model. Although no significant difference in binding of [11C]SMW139 was observed between post mortem brain tissue of Alzheimer's disease patients and that of healthy controls in in vitro autoradiography experiments, [11C]SMW139 could be a promising tracer for P2X7 receptor imaging using positron emission tomography, due to high receptor binding in vivo in the hP2X7 receptor overexpressing rat model. However, further investigation of both P2X7 receptor expression and binding of [11C]SMW139 in other neurological diseases involving microglial activation is warranted.
Collapse
Affiliation(s)
- Bieneke Janssen
- Department of Radiology & Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.
| | - Danielle J Vugts
- Department of Radiology & Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Dieter Ory
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Sylvie Chalon
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Jeroen J M Hoozemans
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Robert C Schuit
- Department of Radiology & Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Wissam Beaino
- Department of Radiology & Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Esther J M Kooijman
- Department of Radiology & Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Johan van den Hoek
- Department of Radiology & Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Mansoor Chishty
- Pharmidex Pharmaceutical Services Ltd., London, United Kingdom
| | - Aurélie Doméné
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Anke Van der Perren
- Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Alessandro Villa
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Ger T Molenaar
- Department of Radiology & Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- BV Cyclotron VU, Amsterdam, The Netherlands
| | - Uta Funke
- Department of Radiology & Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- BV Cyclotron VU, Amsterdam, The Netherlands
| | | | - Veerle Baekelandt
- Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Michael Kassiou
- School of Chemistry, University of Sydney, Sydney, Australia
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
18
|
Wei L, Syed Mortadza SA, Yan J, Zhang L, Wang L, Yin Y, Li C, Chalon S, Emond P, Belzung C, Li D, Lu C, Roger S, Jiang LH. ATP-activated P2X7 receptor in the pathophysiology of mood disorders and as an emerging target for the development of novel antidepressant therapeutics. Neurosci Biobehav Rev 2018; 87:192-205. [PMID: 29453990 DOI: 10.1016/j.neubiorev.2018.02.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/01/2018] [Accepted: 02/08/2018] [Indexed: 12/11/2022]
Abstract
Mood disorders are a group of psychiatric conditions that represent leading global disease burdens. Increasing evidence from clinical and preclinical studies supports that innate immune system dysfunction plays an important part in the pathophysiology of mood disorders. P2X7 receptor, belonging to the ligand-gated ion channel P2X subfamily of purinergic P2 receptors for extracellular ATP, is highly expressed in immune cells including microglia in the central nervous system (CNS) and has a vital role in mediating innate immune response. The P2X7 receptor is also important in neuron-glia signalling in the CNS. The gene encoding human P2X7 receptor is located in a locus of susceptibility to mood disorders. In this review, we will discuss the recent progress in understanding the role of the P2X7 receptor in the pathogenesis and development of mood disorders and in discovering CNS-penetrable P2X7 antagonists for potential uses in in vivo imaging to monitor brain inflammation and antidepressant therapeutics.
Collapse
Affiliation(s)
- Linyu Wei
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, China; School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, United Kingdom
| | - Sharifah A Syed Mortadza
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, United Kingdom; Faculty of Medicine and Health Science, University Putra Malaysia, Selangor, Malaysia
| | - Jing Yan
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, China
| | - Libin Zhang
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, China
| | - Lu Wang
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, China
| | - Yaling Yin
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, China
| | - Chaokun Li
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, China
| | - Sylvie Chalon
- Inserm UMR 1253, iBrain, Université de Tours, Tours, France
| | - Patrick Emond
- Inserm UMR 1253, iBrain, Université de Tours, Tours, France; CHRU de Tours, Service de Médecine Nucléaire In Vitro, Tours, France
| | | | - Dongliang Li
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, China; Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University, China
| | - Chengbiao Lu
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, China; Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University, China
| | - Sebastien Roger
- Inserm UMR1069, Nutrition, Croissance et Cancer, Université de Tours, France; Institut Universitaire de France, Paris Cedex 05, France
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, China; School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, United Kingdom; Institut Universitaire de France, Paris Cedex 05, France.
| |
Collapse
|
19
|
Savio LEB, de Andrade Mello P, da Silva CG, Coutinho-Silva R. The P2X7 Receptor in Inflammatory Diseases: Angel or Demon? Front Pharmacol 2018; 9:52. [PMID: 29467654 PMCID: PMC5808178 DOI: 10.3389/fphar.2018.00052] [Citation(s) in RCA: 299] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/15/2018] [Indexed: 12/13/2022] Open
Abstract
Under physiological conditions, adenosine triphosphate (ATP) is present at low levels in the extracellular milieu, being massively released by stressed or dying cells. Once outside the cells, ATP and related nucleotides/nucleoside generated by ectonucleotidases mediate a high evolutionary conserved signaling system: the purinergic signaling, which is involved in a variety of pathological conditions, including inflammatory diseases. Extracellular ATP has been considered an endogenous adjuvant that can initiate inflammation by acting as a danger signal through the activation of purinergic type 2 receptors-P2 receptors (P2Y G-protein coupled receptors and P2X ligand-gated ion channels). Among the P2 receptors, the P2X7 receptor is the most extensively studied from an immunological perspective, being involved in both innate and adaptive immune responses. P2X7 receptor activation induces large-scale ATP release via its intrinsic ability to form a membrane pore or in association with pannexin hemichannels, boosting purinergic signaling. ATP acting via P2X7 receptor is the second signal to the inflammasome activation, inducing both maturation and release of pro-inflammatory cytokines, such as IL-1β and IL-18, and the production of reactive nitrogen and oxygen species. Furthermore, the P2X7 receptor is involved in caspases activation, as well as in apoptosis induction. During adaptive immune response, P2X7 receptor modulates the balance between the generation of T helper type 17 (Th17) and T regulatory (Treg) lymphocytes. Therefore, this receptor is involved in several inflammatory pathological conditions. In infectious diseases and cancer, P2X7 receptor can have different and contrasting effects, being an angel or a demon depending on its level of activation, cell studied, type of pathogen, and severity of infection. In neuroinflammatory and neurodegenerative diseases, P2X7 upregulation and function appears to contribute to disease progression. In this review, we deeply discuss P2X7 receptor dual function and its pharmacological modulation in the context of different pathologies, and we also highlight the P2X7 receptor as a potential target to treat inflammatory related diseases.
Collapse
Affiliation(s)
- Luiz E B Savio
- Laboratory of Immunophysiology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paola de Andrade Mello
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, United States
| | - Cleide Gonçalves da Silva
- Division of Vascular Surgery, Department of Surgery, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Robson Coutinho-Silva
- Laboratory of Immunophysiology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Bhattacharya A. Recent Advances in CNS P2X7 Physiology and Pharmacology: Focus on Neuropsychiatric Disorders. Front Pharmacol 2018; 9:30. [PMID: 29449810 PMCID: PMC5799703 DOI: 10.3389/fphar.2018.00030] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/10/2018] [Indexed: 12/17/2022] Open
Abstract
The ATP-gated P2X7 ion channel is an abundant microglial protein in the CNS that plays an important pathological role in executing ATP-driven danger signal transduction. Emerging data has generated scientific interest and excitement around targeting the P2X7 ion channel as a potential drug target for CNS disorders. Over the past years, a wealth of data has been published on CNS P2X7 biology, in particular the role of P2X7 in microglial cells, and in vivo effects of brain-penetrant P2X7 antagonists. Likewise, significant progress has been made around the medicinal chemistry of CNS P2X7 ligands, as antagonists for in vivo target validation in models of CNS diseases, to identification of two clinical compounds (JNJ-54175446 and JNJ-55308942) and finally, discovery of P2X7 PET ligands. This review is an attempt to bring together the current understanding of P2X7 in the CNS with a focus on P2X7 as a drug target in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Anindya Bhattacharya
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, CA, United States
| |
Collapse
|
21
|
Zhao H, Chen Y, Feng H. P2X7 Receptor-Associated Programmed Cell Death in the Pathophysiology of Hemorrhagic Stroke. Curr Neuropharmacol 2018; 16:1282-1295. [PMID: 29766811 PMCID: PMC6251042 DOI: 10.2174/1570159x16666180516094500] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 07/17/2017] [Accepted: 02/28/2018] [Indexed: 12/20/2022] Open
Abstract
Hemorrhagic stroke is a life-threatening disease characterized by a sudden rupture of cerebral blood vessels, and cell death is widely believed to occur after exposure to blood metabolites or subsequently damaged cells. Recently, programmed cell death, such as apoptosis, autophagy, necroptosis, pyroptosis, and ferroptosis, has been demonstrated to play crucial roles in the pathophysiology of stroke. However, the detailed mechanisms of these novel kinds of cell death are still unclear. The P2X7 receptor, previously known for its cytotoxic activity, is an ATP-gated, nonselective cation channel that belongs to the family of ionotropic P2X receptors. Evolving evidence indicates that the P2X7 receptor plays a pivotal role in central nervous system pathology; genetic deletion and pharmacological blockade of the P2X7 receptor provide neuroprotection in various neurological disorders, including intracerebral hemorrhage and subarachnoid hemorrhage. The P2X7 receptor may regulate programmed cell death via (I) exocytosis of secretory lysosomes, (II) exocytosis of autophagosomes or autophagolysosomes during formation of the initial autophagic isolation membrane or omegasome, and (III) direct release of cytosolic IL-1β secondary to regulated cell death by pyroptosis or necroptosis. In this review, we present an overview of P2X7 receptor- associated programmed cell death for further understanding of hemorrhagic stroke pathophysiology, as well as potential therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Hengli Zhao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| |
Collapse
|
22
|
Jin H, Han J, Resing D, Liu H, Yue X, Miller RL, Schoch KM, Miller TM, Perlmutter JS, Egan TM, Tu Z. Synthesis and in vitro characterization of a P2X7 radioligand [ 123I]TZ6019 and its response to neuroinflammation in a mouse model of Alzheimer disease. Eur J Pharmacol 2017; 820:8-17. [PMID: 29225193 DOI: 10.1016/j.ejphar.2017.12.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/21/2017] [Accepted: 12/04/2017] [Indexed: 02/08/2023]
Abstract
The purinergic receptor P2X ligand-gated ion channel 7 (P2X7 receptor) is a promising imaging target to detect neuroinflammation. Herein, we report development of a potent iodinated radiotracer for P2X7 receptor, [123I]TZ6019. The radiosynthesis of [123I]TZ6019 was accomplished by allylic-tin precursor iodination using [123I]NaI with good radiochemical yield of 85% and high radiochemical purity of > 99%. Human embryonic kidney 293 (HEK-293) cell line stably transfected with the human P2X7 receptor was used to characterize the binding affinity of TZ6019 by fluorescence, radioactive competitive, and saturation binding assays. A radioligand competitive binding assay with [123I]TZ6019 demonstrated that the nonradioactive compound TZ6019 has an IC50 value of 9.49 ± 1.4nM, and the known P2X7 receptor compound GSK1482160 has an IC50 value of 4.30 ± 0.86nM, consistent with previous reports. The radioligand saturation binding assay and competitive assay revealed that [123I]TZ6019 specifically bound to the human P2X7 receptor with high affinity (Ki = 6.3 ± 0.9nM). In vitro autoradiography quantification with brain slices collected from 9-month old P301S tau transgenic mice along with wild type controls, revealed higher binding of [123I]TZ6019 (35% increase) in the brain of P301S transgenic mice (n = 3, p = 0.04) compared to wild type controls. The immunofluorescence microscopy confirmed that expression of P2X7 receptor was colocalized with astrocytes in the tauopathy P301S transgenic mice. [123I]TZ6019 has specific binding for P2X7 receptor and has great potential to be a radiotracer for screening new compounds and quantifying expression of P2X7 receptor in neuroinflammation related diseases.
Collapse
Affiliation(s)
- Hongjun Jin
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Junbin Han
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Derek Resing
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hui Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xuyi Yue
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rebecca L Miller
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kathleen M Schoch
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Timothy M Miller
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; Neuroscience, Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Terrance M Egan
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
23
|
Gonzaga DTG, Ferreira LBG, Moreira Maramaldo Costa TE, von Ranke NL, Anastácio Furtado Pacheco P, Sposito Simões AP, Arruda JC, Dantas LP, de Freitas HR, de Melo Reis RA, Penido C, Bello ML, Castro HC, Rodrigues CR, Ferreira VF, Faria RX, da Silva FDC. 1-Aryl-1 H - and 2-aryl-2 H -1,2,3-triazole derivatives blockade P2X7 receptor in vitro and inflammatory response in vivo. Eur J Med Chem 2017; 139:698-717. [DOI: 10.1016/j.ejmech.2017.08.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/02/2017] [Accepted: 08/15/2017] [Indexed: 01/09/2023]
|
24
|
Letavic MA, Savall BM, Allison BD, Aluisio L, Andres JI, De Angelis M, Ao H, Beauchamp DA, Bonaventure P, Bryant S, Carruthers NI, Ceusters M, Coe KJ, Dvorak CA, Fraser IC, Gelin CF, Koudriakova T, Liang J, Lord B, Lovenberg TW, Otieno MA, Schoetens F, Swanson DM, Wang Q, Wickenden AD, Bhattacharya A. 4-Methyl-6,7-dihydro-4H-triazolo[4,5-c]pyridine-Based P2X7 Receptor Antagonists: Optimization of Pharmacokinetic Properties Leading to the Identification of a Clinical Candidate. J Med Chem 2017; 60:4559-4572. [PMID: 28493698 DOI: 10.1021/acs.jmedchem.7b00408] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The synthesis and preclinical characterization of novel 4-(R)-methyl-6,7-dihydro-4H-triazolo[4,5-c]pyridines that are potent and selective brain penetrant P2X7 antagonists are described. Optimization efforts based on previously disclosed unsubstituted 6,7-dihydro-4H-triazolo[4,5-c]pyridines, methyl substituted 5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyrazines, and several other series lead to the identification of a series of 4-(R)-methyl-6,7-dihydro-4H-triazolo[4,5-c]pyridines that are selective P2X7 antagonists with potency at the rodent and human P2X7 ion channels. These novel P2X7 antagonists have suitable physicochemical properties, and several analogs have an excellent pharmacokinetic profile, good partitioning into the CNS and show robust in vivo target engagement after oral dosing. Improvements in metabolic stability led to the identification of JNJ-54175446 (14) as a candidate for clinical development. The drug discovery efforts and strategies that resulted in the identification of the clinical candidate are described herein.
Collapse
Affiliation(s)
- Michael A Letavic
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Brad M Savall
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Brett D Allison
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Leah Aluisio
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Jose Ignacio Andres
- Janssen Research & Development, a Division of Janssen-Cilag , Jarama 75, 45007 Toledo, Spain
| | - Meri De Angelis
- Janssen Research & Development, a Division of Janssen-Cilag , Jarama 75, 45007 Toledo, Spain
| | - Hong Ao
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Derek A Beauchamp
- Janssen Research & Development, LLC , 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Pascal Bonaventure
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Stewart Bryant
- Janssen Research & Development, LLC , 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Nicholas I Carruthers
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Marc Ceusters
- Janssen Research & Development, Janssen Pharmaceutica NV , Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Kevin J Coe
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Curt A Dvorak
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Ian C Fraser
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Christine F Gelin
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Tatiana Koudriakova
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Jimmy Liang
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Brian Lord
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Timothy W Lovenberg
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Monicah A Otieno
- Janssen Research & Development, LLC , 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Freddy Schoetens
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Devin M Swanson
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Qi Wang
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Alan D Wickenden
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Anindya Bhattacharya
- Janssen Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| |
Collapse
|
25
|
|
26
|
Abstract
INTRODUCTION The P2X7 receptor (P2X7R) is a unique subtype among the family of seven purinergic P2X receptors, which are ATP-gated non-selective cation channels. P2X7R has been reported to have pathological roles in various diseases, including autoimmune diseases such as arthritis and inflammatory bowel disease, neurodegenerative diseases, chronic pain, mood disorders and cancers. Therefore, many pharmaceutical companies have endeavored to develop a clinical candidate targeting P2X7R. Areas covered: This review provides a summary of various patents on chemicals and biologics and their clinical use published between 2010 and 2015. The reader will gain information on structural claims, representative structures and biological activities of recently developed P2X7R antagonists. Expert opinion: P2X7R is a fascinating therapeutic target and potential biomarker for inflammation, pain disorders and cancers. Research on the development of P2X7R antagonists has continually increased despite the failure of AstraZeneca and Merck's compounds in phase II clinical trials. Various scaffolds have been disclosed by several pharmaceutical industries, and some compounds are currently under investigation in clinical trials.
Collapse
Affiliation(s)
- Jin-Hee Park
- a School of Life Sciences , Gwangju Institute of Science & Technology , Gwangju , Republic of Korea.,b New Drug Development Center (NDDC) , Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF) , Daegu , Republic of Korea
| | - Yong-Chul Kim
- a School of Life Sciences , Gwangju Institute of Science & Technology , Gwangju , Republic of Korea.,c Department of Biomedical Science and Engineering , Gwangju Institute of Science & Technology , Gwangju , Republic of Korea
| |
Collapse
|
27
|
The evolution of P2X7 antagonists with a focus on CNS indications. Bioorg Med Chem Lett 2016; 26:3838-45. [PMID: 27426304 DOI: 10.1016/j.bmcl.2016.06.048] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/08/2016] [Accepted: 06/16/2016] [Indexed: 12/11/2022]
Abstract
The P2X7 receptor is an ATP-gated nonselective cation channel that has been linked to a number of inflammatory diseases. Activation of the P2X7 receptor by elevated levels of ATP results in the release of proinflammatory cytokines and elevated levels of these cytokines has been associated with a variety of disease states. A number of research groups in both industry and academia have explored the identification of P2X7R antagonists as therapeutic agents. Much of this early effort focused on the treatment of diseases related to peripheral inflammation and resulted in several clinical candidates, none of which were advanced to market. The emerging role of the P2X7 receptor in neuroinflammation and related diseases has resulted in a shift in medicinal chemistry efforts toward the development of centrally penetrant antagonists. This review will highlight the biology supporting the role of P2X7 in diseases related to neuroinflammation and review the recent medicinal chemistry efforts to identify centrally penetrant antagonists.
Collapse
|
28
|
Fischer W, Franke H, Krügel U, Müller H, Dinkel K, Lord B, Letavic MA, Henshall DC, Engel T. Critical Evaluation of P2X7 Receptor Antagonists in Selected Seizure Models. PLoS One 2016; 11:e0156468. [PMID: 27281030 PMCID: PMC4900628 DOI: 10.1371/journal.pone.0156468] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 05/13/2016] [Indexed: 01/03/2023] Open
Abstract
The ATP-gated P2X7 receptor (P2X7R) is a non-selective cation channel which senses high extracellular ATP concentrations and has been suggested as a target for the treatment of neuroinflammation and neurodegenerative diseases. The use of P2X7R antagonists may therefore be a viable approach for treating CNS pathologies, including epileptic disorders. Recent studies showed anticonvulsant potential of P2X7R antagonists in certain animal models. To extend this work, we tested three CNS-permeable P2X7R blocker (Brilliant Blue G, AFC-5128, JNJ-47965567) and a natural compound derivative (tanshinone IIA sulfonate) in four well-characterized animal seizure models. In the maximal electroshock seizure threshold test and the pentylenetetrazol (PTZ) seizure threshold test in mice, none of the four compounds demonstrated anticonvulsant effects when given alone. Notably, in combination with carbamazepine, both AFC-5128 and JNJ-47965567 increased the threshold in the maximal electroshock seizure test. In the PTZ-kindling model in rats, useful for testing antiepileptogenic activities, Brilliant Blue G and tanshinone exhibited a moderate retarding effect, whereas the potent P2X7R blocker AFC-5128 and JNJ-47965567 showed a significant and long-lasting delay in kindling development. In fully kindled rats, the investigated compounds revealed modest effects to reduce the mean seizure stage. Furthermore, AFC-5128- and JNJ-47965567-treated animals displayed strongly reduced Iba 1 and GFAP immunoreactivity in the hippocampal CA3 region. In summary, our results show that P2X7R antagonists possess no remarkable anticonvulsant effects in the used acute screening tests, but can attenuate chemically-induced kindling. Further studies would be of interest to support the concept that P2X7R signalling plays a crucial role in the pathogenesis of epileptic disorders.
Collapse
Affiliation(s)
- Wolfgang Fischer
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, Medical Faculty, University of Leipzig, Leipzig, Germany
- * E-mail:
| | - Heike Franke
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Ute Krügel
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | | | - Klaus Dinkel
- Lead Discovery Center GmbH, Dortmund, Germany
- Affectis Pharmaceutical AG, Dortmund, Germany
| | - Brian Lord
- Neuroscience Therapeutic Area, Janssen Research & Development, LLC, San Diego, California, United States of America
| | - Michael A. Letavic
- Neuroscience Therapeutic Area, Janssen Research & Development, LLC, San Diego, California, United States of America
| | - David C. Henshall
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Tobias Engel
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| |
Collapse
|
29
|
Bhattacharya A, Biber K. The microglial ATP-gated ion channel P2X7 as a CNS drug target. Glia 2016; 64:1772-87. [PMID: 27219534 DOI: 10.1002/glia.23001] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 04/17/2016] [Accepted: 04/18/2016] [Indexed: 12/14/2022]
Abstract
Based on promising preclinical evidence, microglial P2X7 has increasingly being recognized as a target for therapeutic intervention in neurological and psychiatric diseases. However, despite this knowledge no P2X7-related drug has yet entered clinical trials with respect to CNS diseases. We here discuss the current literature on P2X7 being a drug target and identify unsolved issues and still open questions that have hampered the development of P2X7 dependent therapeutic approaches for CNS diseases. It is concluded here that the lack of brain penetrating P2X7 antagonists is a major obstacle in the field and that central P2X7 is a yet untested clinical drug target. In the CNS, microglial P2X7 activation causes neuroinflammation, which in turn plays a role in various CNS disorders. This has resulted in a surge of brain penetrant P2X7 antagonists. P2X7 is a viable, clinically untested CNS drug target. GLIA 2016;64:1772-1787.
Collapse
Affiliation(s)
- Anindya Bhattacharya
- LLC. Neuroscience Drug Discovery, Janssen Research & Development, 3210 Merryfield Row, San Diego, California
| | - Knut Biber
- Department of Psychiatry and Psychotherapy, University Hospital Freiburg, Hauptstrasse 5, Freiburg, Germany.,Department of Neuroscience, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, AV Groningen, The Netherlands
| |
Collapse
|
30
|
Ziff J, Rudolph DA, Stenne B, Koudriakova T, Lord B, Bonaventure P, Lovenberg TW, Carruthers NI, Bhattacharya A, Letavic MA, Shireman BT. Substituted 5,6-(Dihydropyrido[3,4-d]pyrimidin-7(8H)-yl)-methanones as P2X7 Antagonists. ACS Chem Neurosci 2016; 7:498-504. [PMID: 26754558 DOI: 10.1021/acschemneuro.5b00304] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We describe the synthesis of a novel class of brain penetrating P2X7 antagonists with high potency at both the rat and human P2X7 receptors. Disclosed herein are druglike molecules with demonstrated target engagement of the rat P2X7 receptors after an oral dose. Specifically, compound 20 occupied the P2X7 receptors >80% over the 6 h time course as measured by an ex vivo radioligand binding experiment. In a dose-response assay, this molecule has a plasma EC50 of 8 ng/mL. Overall, 20 has suitable druglike properties and pharmacokinetics in rat and dog. This molecule and others disclosed herein will serve as additional tools to elucidate the role of the P2X7 receptor in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jeannie Ziff
- Janssen Research & Development L.L.C., 3210 Merryfield Row, San Diego, California 92121, United States
| | - Dale A. Rudolph
- Janssen Research & Development L.L.C., 3210 Merryfield Row, San Diego, California 92121, United States
| | - Brice Stenne
- Janssen Research & Development L.L.C., 3210 Merryfield Row, San Diego, California 92121, United States
| | - Tatiana Koudriakova
- Janssen Research & Development L.L.C., 3210 Merryfield Row, San Diego, California 92121, United States
| | - Brian Lord
- Janssen Research & Development L.L.C., 3210 Merryfield Row, San Diego, California 92121, United States
| | - Pascal Bonaventure
- Janssen Research & Development L.L.C., 3210 Merryfield Row, San Diego, California 92121, United States
| | - Timothy W. Lovenberg
- Janssen Research & Development L.L.C., 3210 Merryfield Row, San Diego, California 92121, United States
| | - Nicholas I. Carruthers
- Janssen Research & Development L.L.C., 3210 Merryfield Row, San Diego, California 92121, United States
| | - Anindya Bhattacharya
- Janssen Research & Development L.L.C., 3210 Merryfield Row, San Diego, California 92121, United States
| | - Michael A. Letavic
- Janssen Research & Development L.L.C., 3210 Merryfield Row, San Diego, California 92121, United States
| | - Brock T. Shireman
- Janssen Research & Development L.L.C., 3210 Merryfield Row, San Diego, California 92121, United States
| |
Collapse
|
31
|
Jacobson KA, Müller CE. Medicinal chemistry of adenosine, P2Y and P2X receptors. Neuropharmacology 2015; 104:31-49. [PMID: 26686393 DOI: 10.1016/j.neuropharm.2015.12.001] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/30/2015] [Accepted: 12/01/2015] [Indexed: 12/13/2022]
Abstract
Pharmacological tool compounds are now available to define action at the adenosine (ARs), P2Y and P2X receptors. We present a selection of the most commonly used agents to study purines in the nervous system. Some of these compounds, including A1 and A3 AR agonists, P2Y1R and P2Y12R antagonists, and P2X3, P2X4 and P2X7 antagonists, are potentially of clinical use in treatment of disorders of the nervous system, such as chronic pain, neurodegeneration and brain injury. Agonists of the A2AAR and P2Y2R are already used clinically, P2Y12R antagonists are widely used antithrombotics and an antagonist of the A2AAR is approved in Japan for treating Parkinson's disease. The selectivity defined for some of the previously introduced compounds has been revised with updated pharmacological characterization, for example, various AR agonists and antagonists were deemed A1AR or A3AR selective based on human data, but species differences indicated a reduction in selectivity ratios in other species. Also, many of the P2R ligands still lack bioavailability due to charged groups or hydrolytic (either enzymatic or chemical) instability. X-ray crystallographic structures of AR and P2YRs have shifted the mode of ligand discovery to structure-based approaches rather than previous empirical approaches. The X-ray structures can be utilized either for in silico screening of chemically diverse libraries for the discovery of novel ligands or for enhancement of the properties of known ligands by chemical modification. Although X-ray structures of the zebrafish P2X4R have been reported, there is scant structural information about ligand recognition in these trimeric ion channels. In summary, there are definitive, selective agonists and antagonists for all of the ARs and some of the P2YRs; while the pharmacochemistry of P2XRs is still in nascent stages. The therapeutic potential of selectively modulating these receptors is continuing to gain interest in such fields as cancer, inflammation, pain, diabetes, ischemic protection and many other conditions. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Key Words
- 2-MeSADP, (PubChem CID: 121990)
- A-740003, (PubChem CID: 23232014)
- ATP
- Agonists
- Antagonists
- DPCPX, (PubChem CID: 1329)
- GPCR
- IB-MECA, (PubChem CID: 123683)
- Ion channel
- LUF6000, (PubChem CID: 11711282)
- MRS2500, (PubChem CID: 44448831)
- Nucleosides
- Nucleotides
- PPTN, (PubChem CID: 42611190)
- PSB-1114, (PubChem CID: 52952605)
- PSB-603, (PubChem CID: 44185871)
- SCH442416, (PubChem CID: 10668061)
Collapse
Affiliation(s)
- Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 20892, Bethesda, USA.
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany
| |
Collapse
|
32
|
Lord B, Ameriks MK, Wang Q, Fourgeaud L, Vliegen M, Verluyten W, Haspeslagh P, Carruthers NI, Lovenberg TW, Bonaventure P, Letavic MA, Bhattacharya A. A novel radioligand for the ATP-gated ion channel P2X7: [3H] JNJ-54232334. Eur J Pharmacol 2015; 765:551-9. [PMID: 26386289 DOI: 10.1016/j.ejphar.2015.09.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 09/14/2015] [Accepted: 09/15/2015] [Indexed: 10/25/2022]
Abstract
The ATP-gated ion channel P2X7 has emerged as a potential central nervous system (CNS) drug target based on the hypotheses that pro-inflammatory cytokines such as IL-1β that are released by microglia, may contribute to the etiology of various disorders of the CNS including depression. In this study, we identified two closely related P2X7 antagonists, JNJ-54232334 and JNJ-54140515, and then tritium labeled the former to produce a new radioligand for P2X7. JNJ-54232334 is a high affinity ligand for the rat P2X7 with a pKi of 9.3±0.1. In rat cortical membranes, [3H] JNJ-54232334 reached saturable binding with equilibrium dissociation (Kd) constant of 4.9±1.3 nM. The compound displayed monophasic association and dissociation kinetics with fast on and off rates. In rat brain sections, specific binding of [3H] JNJ-54232334 was markedly improved compared to the previously described P2X7 radioligand, [3H] A-804598. In P2X7 knockout mouse brain sections, [3H] A-804598 bound to non-P2X7 binding sites in contrast to [3H] JNJ-54232334. In rat or wild type mouse brain sections [3H] JNJ-54232334 bound in a more homogenous and region independent manner. The ubiquitous expression of P2X7 receptors was confirmed with immunohistochemistry in rat brain sections. The partial displacement of [3H] A-804598 binding resulted in the underestimation of the level of ex vivo P2X7 occupancy for JNJ-54140515. Higher levels of P2X7 ex vivo occupancy were measured using [3H] JNJ-54232334 due to less non-specific binding. In summary, we describe [3H] JNJ-54232334 as a novel P2X7 radioligand, with improved properties over [3H] A-804598.
Collapse
Affiliation(s)
- Brian Lord
- Janssen Research & Development, LLC, Neuroscience Drug Discovery, 3210 Merryfield Row, San Diego, CA 92121-1126, United States.
| | - Michael K Ameriks
- Janssen Research & Development, LLC, Neuroscience Drug Discovery, 3210 Merryfield Row, San Diego, CA 92121-1126, United States
| | - Qi Wang
- Janssen Research & Development, LLC, Neuroscience Drug Discovery, 3210 Merryfield Row, San Diego, CA 92121-1126, United States
| | - Lawrence Fourgeaud
- Janssen Research & Development, LLC, Neuroscience Drug Discovery, 3210 Merryfield Row, San Diego, CA 92121-1126, United States
| | - Maarten Vliegen
- Janssen Research & Development, LLC, Drug Safety Sciences, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Willy Verluyten
- Janssen Research & Development, LLC, Drug Safety Sciences, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Pieter Haspeslagh
- Janssen Research & Development, LLC, Drug Safety Sciences, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Nicholas I Carruthers
- Janssen Research & Development, LLC, Neuroscience Drug Discovery, 3210 Merryfield Row, San Diego, CA 92121-1126, United States
| | - Timothy W Lovenberg
- Janssen Research & Development, LLC, Neuroscience Drug Discovery, 3210 Merryfield Row, San Diego, CA 92121-1126, United States
| | - Pascal Bonaventure
- Janssen Research & Development, LLC, Neuroscience Drug Discovery, 3210 Merryfield Row, San Diego, CA 92121-1126, United States
| | - Michael A Letavic
- Janssen Research & Development, LLC, Neuroscience Drug Discovery, 3210 Merryfield Row, San Diego, CA 92121-1126, United States
| | - Anindya Bhattacharya
- Janssen Research & Development, LLC, Neuroscience Drug Discovery, 3210 Merryfield Row, San Diego, CA 92121-1126, United States
| |
Collapse
|
33
|
Savall BM, Wu D, De Angelis M, Carruthers NI, Ao H, Wang Q, Lord B, Bhattacharya A, Letavic MA. Synthesis, SAR, and Pharmacological Characterization of Brain Penetrant P2X7 Receptor Antagonists. ACS Med Chem Lett 2015; 6:671-6. [PMID: 26101572 DOI: 10.1021/acsmedchemlett.5b00089] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/24/2015] [Indexed: 01/22/2023] Open
Abstract
We describe the synthesis and SAR of 1,2,3-triazolopiperidines as a novel series of potent, brain penetrant P2X7 antagonists. Initial efforts yielded a series of potent human P2X7R antagonists with moderate to weak rodent potency, some CYP inhibition, poor metabolic stability, and low solubility. Further work in this series, which focused on the SAR of the N-linked heterocycle, not only increased the potency at the human P2X7R but also provided compounds with good potency at the rat P2X7R. These efforts eventually delivered a potent rat and human P2X7R antagonist with good physicochemical properties, an excellent pharmacokinetic profile, good partitioning into the CNS, and demonstrated in vivo target engagement after oral dosing.
Collapse
Affiliation(s)
- Brad M. Savall
- Janssen Research & Development, LLC, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Duncan Wu
- Janssen Research & Development, LLC, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Meri De Angelis
- Janssen Research & Development, Discovery Sciences, a division of Janssen-Cilag, Jarama 75, 45007 Toledo, Spain
| | - Nicholas I. Carruthers
- Janssen Research & Development, LLC, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Hong Ao
- Janssen Research & Development, LLC, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Qi Wang
- Janssen Research & Development, LLC, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Brian Lord
- Janssen Research & Development, LLC, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Anindya Bhattacharya
- Janssen Research & Development, LLC, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Michael A. Letavic
- Janssen Research & Development, LLC, 3210 Merryfield Row, San Diego, California 92121, United States
| |
Collapse
|
34
|
Rudolph DA, Alcazar J, Ameriks MK, Anton AB, Ao H, Bonaventure P, Carruthers NI, Chrovian CC, De Angelis M, Lord B, Rech JC, Wang Q, Bhattacharya A, Andres JI, Letavic MA. Novel methyl substituted 1-(5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl)methanones are P2X7 antagonists. Bioorg Med Chem Lett 2015; 25:3157-63. [PMID: 26099534 DOI: 10.1016/j.bmcl.2015.06.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 05/29/2015] [Accepted: 06/02/2015] [Indexed: 12/25/2022]
Abstract
The optimization efforts that led to a novel series of methyl substituted 1-(5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl)methanones that are potent rat and human P2X7 antagonists are described. These efforts resulted in the discovery of compounds with good drug-like properties that are capable of high P2X7 receptor occupancy in rat following oral administration, including compounds 7n (P2X7 IC50 = 7.7 nM) and 7u (P2X7 IC50 =7 .7 nM). These compounds are expected to be useful tools for characterizing the effects of P2X7 antagonism in models of depression and epilepsy, and several of the compounds prepared are candidates for effective P2X7 PET tracers.
Collapse
Affiliation(s)
- Dale A Rudolph
- Janssen Pharmaceutical Research & Development L.L.C., 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Jesus Alcazar
- Janssen Research & Development, Discovery Sciences, A Division of Janssen-Cilag, Jarama 75, 45007 Toledo, Spain
| | - Michael K Ameriks
- Janssen Pharmaceutical Research & Development L.L.C., 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Ana Belen Anton
- Janssen Research & Development, Discovery Sciences, A Division of Janssen-Cilag, Jarama 75, 45007 Toledo, Spain
| | - Hong Ao
- Janssen Pharmaceutical Research & Development L.L.C., 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Pascal Bonaventure
- Janssen Pharmaceutical Research & Development L.L.C., 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Nicholas I Carruthers
- Janssen Pharmaceutical Research & Development L.L.C., 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Christa C Chrovian
- Janssen Pharmaceutical Research & Development L.L.C., 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Meri De Angelis
- Janssen Research & Development, Discovery Sciences, A Division of Janssen-Cilag, Jarama 75, 45007 Toledo, Spain
| | - Brian Lord
- Janssen Pharmaceutical Research & Development L.L.C., 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Jason C Rech
- Janssen Pharmaceutical Research & Development L.L.C., 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Qi Wang
- Janssen Pharmaceutical Research & Development L.L.C., 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Anindya Bhattacharya
- Janssen Pharmaceutical Research & Development L.L.C., 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Jose Ignacio Andres
- Janssen Research & Development, Discovery Sciences, A Division of Janssen-Cilag, Jarama 75, 45007 Toledo, Spain
| | - Michael A Letavic
- Janssen Pharmaceutical Research & Development L.L.C., 3210 Merryfield Row, San Diego, CA 92121, United States.
| |
Collapse
|
35
|
Bartlett R, Stokes L, Sluyter R. The P2X7 receptor channel: recent developments and the use of P2X7 antagonists in models of disease. Pharmacol Rev 2015; 66:638-75. [PMID: 24928329 DOI: 10.1124/pr.113.008003] [Citation(s) in RCA: 320] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The P2X7 receptor is a trimeric ATP-gated cation channel found predominantly, but not exclusively, on immune cells. P2X7 activation results in a number of downstream events, including the release of proinflammatory mediators and cell death and proliferation. As such, P2X7 plays important roles in various inflammatory, immune, neurologic and musculoskeletal disorders. This review focuses on the use of P2X7 antagonists in rodent models of neurologic disease and injury, inflammation, and musculoskeletal and other disorders. The cloning and characterization of human, rat, mouse, guinea pig, dog, and Rhesus macaque P2X7, as well as recent observations regarding the gating and permeability of P2X7, are discussed. Furthermore, this review discusses polymorphic and splice variants of P2X7, as well as the generation and use of P2X7 knockout mice. Recent evidence for emerging signaling pathways downstream of P2X7 activation and the growing list of negative and positive modulators of P2X7 activation and expression are also described. In addition, the use of P2X7 antagonists in numerous rodent models of disease is extensively summarized. Finally, the use of P2X7 antagonists in clinical trials in humans and future directions exploring P2X7 as a therapeutic target are described.
Collapse
Affiliation(s)
- Rachael Bartlett
- School of Biological Sciences, University of Wollongong, New South Wales, Australia and Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia (R.B., R.S.); and Health Innovations Research Institute, School of Medical Sciences, RMIT University, Bundoora, Victoria, Australia (L.S.)
| | - Leanne Stokes
- School of Biological Sciences, University of Wollongong, New South Wales, Australia and Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia (R.B., R.S.); and Health Innovations Research Institute, School of Medical Sciences, RMIT University, Bundoora, Victoria, Australia (L.S.)
| | - Ronald Sluyter
- School of Biological Sciences, University of Wollongong, New South Wales, Australia and Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia (R.B., R.S.); and Health Innovations Research Institute, School of Medical Sciences, RMIT University, Bundoora, Victoria, Australia (L.S.)
| |
Collapse
|
36
|
Bhattacharya A, Wang Q, Ao H, Shoblock JR, Lord B, Aluisio L, Fraser I, Nepomuceno D, Neff RA, Welty N, Lovenberg TW, Bonaventure P, Wickenden AD, Letavic MA. Pharmacological characterization of a novel centrally permeable P2X7 receptor antagonist: JNJ-47965567. Br J Pharmacol 2014; 170:624-40. [PMID: 23889535 DOI: 10.1111/bph.12314] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 07/08/2013] [Accepted: 07/19/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE An increasing body of evidence suggests that the purinergic receptor P2X, ligand-gated ion channel, 7 (P2X7) in the CNS may play a key role in neuropsychiatry, neurodegeneration and chronic pain. In this study, we characterized JNJ-47965567, a centrally permeable, high-affinity, selective P2X7 antagonist. EXPERIMENTAL APPROACH We have used a combination of in vitro assays (calcium flux, radioligand binding, electrophysiology, IL-1β release) in both recombinant and native systems. Target engagement of JNJ-47965567 was demonstrated by ex vivo receptor binding autoradiography and in vivo blockade of Bz-ATP induced IL-1β release in the rat brain. Finally, the efficacy of JNJ-47965567 was tested in standard models of depression, mania and neuropathic pain. KEY RESULTS JNJ-47965567 is potent high affinity (pKi 7.9 ± 0.07), selective human P2X7 antagonist, with no significant observed speciation. In native systems, the potency of the compound to attenuate IL-1β release was 6.7 ± 0.07 (human blood), 7.5 ± 0.07 (human monocytes) and 7.1 ± 0.1 (rat microglia). JNJ-47965567 exhibited target engagement in rat brain, with a brain EC50 of 78 ± 19 ng·mL(-1) (P2X7 receptor autoradiography) and functional block of Bz-ATP induced IL-1β release. JNJ-47965567 (30 mg·kg(-1) ) attenuated amphetamine-induced hyperactivity and exhibited modest, yet significant efficacy in the rat model of neuropathic pain. No efficacy was observed in forced swim test. CONCLUSION AND IMPLICATIONS JNJ-47965567 is centrally permeable, high affinity P2X7 antagonist that can be used to probe the role of central P2X7 in rodent models of CNS pathophysiology.
Collapse
Affiliation(s)
- Anindya Bhattacharya
- Neuroscience Therapeutic Area, Janssen Pharmaceutical Companies of Johnson & Johnson, San Diego, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Chrovian CC, Rech JC, Bhattacharya A, Letavic MA. P2X7 antagonists as potential therapeutic agents for the treatment of CNS disorders. PROGRESS IN MEDICINAL CHEMISTRY 2014; 53:65-100. [PMID: 24418608 DOI: 10.1016/b978-0-444-63380-4.00002-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The use of P2X7 antagonists to treat inflammatory disorders has garnered considerable interest in recent years. An increasing number of literature reports support the role of P2X7 in inflammatory pathways of the peripheral and central nervous systems (CNSs). A number of CNS indications such as neuropsychiatric and neurodegenerative disorders and neuropathic pain have been linked to a neuroinflammatory response, and clinical studies have shown that inflammatory biomarkers can be mitigated by modulating P2X7. Recent scientific and patent literature describing novel P2X7 antagonists has indicated their use in CNS disorders. In addition, several reports have disclosed the results of administering P2X7 antagonists in pre-clinical models of CNS disease or investigating brain uptake. This review describes small molecule P2X7 antagonists that have first appeared in the literature since 2009 and have potential therapeutic utility in the CNS, or for which new data have emerged implicating their use in CNS indications.
Collapse
Affiliation(s)
| | - Jason C Rech
- Janssen Research and Development, LLC, San Diego, CA, USA
| | | | | |
Collapse
|