1
|
Recourt K, van der Aart J, Jacobs G, de Kam M, Drevets W, van Nueten L, Kanhai K, Siebenga P, Zuiker R, Ravenstijn P, Timmers M, van Gerven J, de Boer P. Characterisation of the pharmacodynamic effects of the P2X7 receptor antagonist JNJ-54175446 using an oral dexamphetamine challenge model in healthy males in a randomised, double-blind, placebo-controlled, multiple ascending dose trial. J Psychopharmacol 2020; 34:1030-1042. [PMID: 32248747 DOI: 10.1177/0269881120914206] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND This is the first report of the pharmacodynamic (PD) effects of the selective, potent and brain-penetrant P2X7 receptor (P2X7R) antagonist JNJ-54175446. Activation of the P2X7R, an adenosine triphosphate-gated ion channel, leads to the production of pro-inflammatory cytokines, which have been linked to neuroinflammation and play a role in the pathogenesis of mood disorders. Previous clinical studies with JNJ-54175446 demonstrated peripheral target engagement of JNJ-54175446 by assessing ex vivo lipopolysaccharide (LPS)-stimulated cytokine production. Blood-brain barrier penetration and a clear dose-receptor occupancy relationship was demonstrated using positron emission tomography. AIMS The objectives of this double-blind, placebo-controlled, translational study were to assess the safety and tolerability of administering multiple doses of JNJ-54175446 and to explore its PD effects using a dexamphetamine challenge. METHODS Subjects (N = 64) were randomised to either JNJ-54175446 (50-450 mg; n = 48) or placebo (n = 16) and underwent a baseline oral 20 mg dexamphetamine challenge followed by 11 consecutive days q.d. dosing with JNJ-54175446/placebo and a randomised crossover dexamphetamine/placebo challenge. RESULTS At all doses tested, JNJ-54175446 was well tolerated and suppressed the ex vivo LPS-induced release of cytokines. At doses ⩾100 mg, JNJ-54175446 attenuated dexamphetamine-induced increases in locomotion and enhanced the mood-elevating effects of dexamphetamine, suggesting that a dose that is approximately twice as high is needed to obtain a central PD response compared to the dose needed for maximum peripheral occupancy. CONCLUSION Overall, the observed pharmacological profile of JNJ-54175446 in the dexamphetamine challenge paradigm is compatible with a potential mood-modulating effect.
Collapse
Affiliation(s)
- Kasper Recourt
- Centre for Human Drug Research, Leiden, The Netherlands.,Leiden University Medical Center, Leiden, The Netherlands
| | | | - Gabriel Jacobs
- Centre for Human Drug Research, Leiden, The Netherlands.,Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Luc van Nueten
- Janssen Research and Development, a Division of Janssen Pharmaceutica N.V., Beerse, Belgium
| | - Kawita Kanhai
- Centre for Human Drug Research, Leiden, The Netherlands
| | | | - Rob Zuiker
- Centre for Human Drug Research, Leiden, The Netherlands
| | - Paulien Ravenstijn
- Janssen Research and Development, a Division of Janssen Pharmaceutica N.V., Beerse, Belgium
| | - Maarten Timmers
- Janssen Research and Development, a Division of Janssen Pharmaceutica N.V., Beerse, Belgium
| | - Joop van Gerven
- Centre for Human Drug Research, Leiden, The Netherlands.,Leiden University Medical Center, Leiden, The Netherlands
| | - Peter de Boer
- Janssen Research and Development, a Division of Janssen Pharmaceutica N.V., Beerse, Belgium
| |
Collapse
|
2
|
Timmers M, Ravenstijn P, Xi L, Triana-Baltzer G, Furey M, Van Hemelryck S, Biewenga J, Ceusters M, Bhattacharya A, van den Boer M, van Nueten L, de Boer P. Clinical pharmacokinetics, pharmacodynamics, safety, and tolerability of JNJ-54175446, a brain permeable P2X7 antagonist, in a randomised single-ascending dose study in healthy participants. J Psychopharmacol 2018; 32:1341-1350. [PMID: 30260294 DOI: 10.1177/0269881118800067] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Central nervous system-derived interleukin-1β plays a role in mood disorders. P2X7 receptor activation by adenosine-triphosphate leads to the release of interleukin-1β. AIMS This first-in-human study evaluated safety, tolerability, pharmacokinetics and pharmacodynamics of a novel central nervous system-penetrant P2X7 receptor antagonist, JNJ-54175446, in healthy participants. METHODS The study had three parts: an ascending-dose study in fasted participants (0.5-300 mg JNJ-54175446); an ascending-dose study in fed participants (50-600 mg); and a cerebrospinal fluid study (300 mg). Target plasma concentrations were based on estimated plasma effective concentration (EC)50 (105 ng/mL) and EC90 (900 ng/mL) values for central nervous system P2X7 receptor binding. RESULTS Seventy-seven participants received a single oral dose of JNJ-54175446 ( n=59) or placebo ( n=18). Area under the curve of concentration time extrapolated to infinity (AUC∞) increased dose-proportionally; maximum concentration (Cmax) of plasma (Cmax,plasma) increased less than dose-proportionally following single doses of JNJ-54175446. Because food increases bioavailability of JNJ-54175446, higher doses were given with food to evaluate safety at higher exposures. The highest Cmax,plasma reached (600 mg, fed) was 1475±163 ng/mL. JNJ-54175446 Cmax in cerebrospinal fluid, a proxy for brain penetration, was seven times lower than in total plasma; unbound Cmax,plasma and Cmax,CSF were comparable (88.3±35.7 vs 114±39 ng/mL). JNJ-54175446 inhibited lipopolysaccharide/3'-O-(4-benzoylbenzoyl)-ATP-induced interleukin-1β release from peripheral blood in a dose-dependent manner (inhibitory concentration (IC)50:82 ng/mL; 95% confidence interval: 48-94). Thirty-three of 59 (55.9%) participants reported at least one treatment-emergent adverse event; the most common adverse event being headache (11/59, 18.6%). CONCLUSION Plasma exposure of JNJ-54175446 was dose-dependent. No serious adverse events occurred. Single-dose administration of JNJ-54175446>10 mg attenuated ex-vivo lipopolysaccharide-induced interleukin-1β release in peripheral blood. Passive brain penetration of JNJ-54175446 was confirmed.
Collapse
Affiliation(s)
- Maarten Timmers
- 1 Janssen Research and Development, Beerse, Belgium.,2 Reference Centre for Biological Markers of Dementia (BIODEM), University of Antwerp, Antwerp, Belgium
| | | | - Liwen Xi
- 3 Janssen Research and Development, Malvern, PA, USA
| | | | - Maura Furey
- 4 Janssen Research and Development, San Diego, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Danino O, Grossman S, Fischer B. ATP-γ-S-(α,β-CH2) protects against oxidative stress and amyloid beta toxicity in neuronal culture. Biochem Biophys Res Commun 2015; 460:446-50. [PMID: 25796332 DOI: 10.1016/j.bbrc.2015.03.053] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 03/10/2015] [Indexed: 10/23/2022]
Abstract
Amyloid beta (Aβ) oligomers and oxidative stress, typical of Alzheimer's disease, are highly neurotoxic. Previously we identified ATP-γ-S as a most promising antioxidant and neuroprotectant. To further improve both potency and metabolic stability of ATP-γ-S, we designed a related analogue, ATP-γ-S-(α,β-CH2). We found that ATP-γ-S-(α,β-CH2) effectively inhibited ROS formation in PC12 cells subjected to Fe(II)-oxidation, slightly better than ATP-γ-S (IC50 0.18 and 0.20 μM, respectively). Moreover, ATP-γ-S-(α,β-CH2) rescued primary neurons from Aβ42 toxicity, 4-fold more potently than ATP-γ-S, (IC50 0.2 and 0.8 μM, respectively). In addition, the metabolic stability of ATP-γ-S-(α,β-CH2) in PC12 cells during 4 h of incubation, was up to 20% greater than that of ATP-γ-S and ATP. Previously, we found that ATP-γ-S-(α,β-CH2) resisted hydrolysis by ecto-nucleotidases such as, NPPs and TNAP, and was found to be ∼7-fold more potent agonist than ATP at P2Y11 receptor. Therefore, we propose ATP-γ-S-(α,β-CH2) as a promising agent for rescue of neurons from insults typical of Alzheimer's disease.
Collapse
Affiliation(s)
- Ortal Danino
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 52900, Israel.
| | - Shlomo Grossman
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel.
| | - Bilha Fischer
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 52900, Israel.
| |
Collapse
|
4
|
Abstract
Chronic inflammation and oxidative stress have been implicated in the pathophysiology of Major Depressive Disorder (MDD), as well as in a number of chronic medical conditions. The aim of this study was to examine the relationship between peripheral inflammatory and oxidative stress markers in un-medicated subjects with MDD compared to non-depressed healthy controls and compared to subjects with MDD after antidepressant treatment. We examined the relationships between IL-6, IL-10, and the IL-6/IL-10 inflammatory ratio vs. F2-isoprostanes (F2-IsoP), a marker of oxidative stress, in un-medicated MDD patients (n=20) before and after 8 weeks of open-label sertraline treatment (n=17), compared to healthy non-depressed controls (n=20). Among the un-medicated MDD subjects, F2-IsoP concentrations were positively correlated with IL-6 concentrations (p<0.05) and were negatively correlated with IL-10 concentrations (p<0.01). Accordingly, F2-IsoP concentrations were positively correlated with the ratio of IL-6/IL-10 (p<0.01). In contrast, in the control group, there were no significant correlations between F2-IsoPs and either cytokine or their ratio. After MDD subjects were treated with sertraline for 8 weeks, F2-IsoPs were no longer significantly correlated with IL-6, IL-10 or the IL-6/IL-10 ratio. These data suggest oxidative stress and inflammatory processes are positively associated in untreated MDD. Our findings are consistent with the hypothesis that the homeostatic buffering mechanisms regulating oxidation and inflammation in healthy individuals become dysregulated in untreated MDD, and may be improved with antidepressant treatment. These findings may help explain the increased risk of comorbid medical illnesses in MDD.
Collapse
|
5
|
Wesselius A, Bours MJL, Arts ICW, Theunisz EHE, Geusens P, Dagnelie PC. The P2X(7) loss-of-function Glu496Ala polymorphism affects ex vivo cytokine release and protects against the cytotoxic effects of high ATP-levels. BMC Immunol 2012; 13:64. [PMID: 23210974 PMCID: PMC3526505 DOI: 10.1186/1471-2172-13-64] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 11/23/2012] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The P2X(7) receptor plays an important role in cytokine release during the inflammatory response in vivo. Polymorphisms within the P2X(7) receptor gene that lead to loss of receptor function may contribute to impaired cytokine release by immune cells. Therefore, we investigated whether a known loss-of-function polymorphism (Glu496Ala) in the P2X(7) receptor gene leads to alterations in cytokine release in response to ATP. RESULTS An ex vivo whole blood model was used to induce an inflammatory reaction with the pro-inflammatory stimuli LPS and PHA (phytohemagglutinin). Blood from n=9 subjects with the Glu496Ala P2X7 SNP (P2X7MUT) and n=7 'wild-type' subjects (no P2X7 SNP; P2X7WT) was used.Addition of ATP (0.9-3 mM) to LPS/PHA-stimulated whole blood induced an increase in IL-1β release in P2X7MUT subjects, whereas decreased release was observed in P2X7WT subjects. Decreased levels of IL-6 and TNF-α in response to ATP were shown in both P2X7MUT and P2X7WT subjects, which was less pronounced in P2X7MUT subjects. ATP at 3 mM also significantly decreased levels of lactate dehydrogenase (LDH) in P2X7MUT subjects compared to P2X7WT subjects. CONCLUSIONS The presence of the non-synonymous Glu496Ala loss-of-function polymorphism within the P2X(7) receptor gene is likely to be of importance in the release of cytokines during inflammation. Furthermore, this study suggests that carriers of the Glu496Ala loss-of-function polymorphism are protected against the cytotoxic effects of high ATP-levels.
Collapse
Affiliation(s)
- Anke Wesselius
- Department of Epidemiology, Maastricht University, School for Public Health and Primary Care (CAPHRI), Peter Debyeplein 1, Maastricht, MD 6200, The Netherlands.
| | | | | | | | | | | |
Collapse
|
6
|
Swennen ELR, Dagnelie PC, Van den Beucken T, Bast A. Radioprotective effects of ATP in human blood ex vivo. Biochem Biophys Res Commun 2007; 367:383-7. [PMID: 18164682 DOI: 10.1016/j.bbrc.2007.12.125] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2007] [Accepted: 12/20/2007] [Indexed: 11/19/2022]
Abstract
Damage to healthy tissue is a major limitation of radiotherapy treatment of cancer patients, leading to several side effects and complications. Radiation-induced release of pro-inflammatory cytokines is thought to be partially responsible for the radiation-associated complications. The aim of the present study was to investigate the protective effects of extracellular ATP on markers of oxidative stress, radiation-induced inflammation and DNA damage in irradiated blood ex vivo. ATP inhibited radiation-induced TNF-alpha release and increased IL-10 release. The inhibitory effect of ATP on TNF- alpha release was completely reversed by adenosine 5'-O-thiomonophosphate, indicating a P2Y(11) mediated effect. Furthermore, ATP attenuated radiation-induced DNA damage immediate, 3 and 6h after irradiation. Our study indicates that ATP administration alleviates radiation-toxicity to blood cells, mainly by inhibiting radiation-induced inflammation and DNA damage.
Collapse
Affiliation(s)
- Els L R Swennen
- Department of Epidemiology, NUTRIM Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | | | | | | |
Collapse
|
7
|
Bours MJ, Troost FJ, Brummer RJM, Bast A, Dagnelie PC. Local effect of adenosine 5'-triphosphate on indomethacin-induced permeability changes in the human small intestine. Eur J Gastroenterol Hepatol 2007; 19:245-50. [PMID: 17301652 DOI: 10.1097/meg.0b013e328011093c] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Nonsteroidal anti-inflammatory drug (NSAID) use is associated with an elevated risk of gastrointestinal damage. As adenosine 5'-triphosphate (ATP) may play a protective role in the small intestine, our objective was to determine the local effect of ATP on small intestinal permeability changes induced by short-term challenge of the NSAID indomethacin in healthy humans. METHODS Mucosal permeability of the small intestine was assessed by the lactulose/rhamnose permeability test, that is, ingestion of a test drink containing 5 g lactulose and 0.5 g L-rhamnose followed by total urine collection for 5 h. Urinary excretion of lactulose and L-rhamnose was determined by fluorescent detection high-pressure liquid chromatography (HPLC). Basal small intestinal permeability was assessed as a control condition. As a model of increased small intestinal permeability, two doses of indomethacin were ingested before ingestion of the test drink (75 mg and 50 mg at 10 h and 1 h before the test drink, respectively). Concomitantly with indomethacin ingestion, placebo or 30 mg/kg ATP was administered through a naso-intestinal tube. RESULTS Median urinary lactulose/rhamnose ratio (g/g) in the control condition was 0.023 (interquartile range: 0.013-0.041). Compared with the control condition, urinary lactulose/rhamnose ratio after ingestion of indomethacin and administration of placebo was significantly increased [0.042 (0.028-0.076); P<0.01]. In contrast, urinary lactulose/rhamnose ratio after indomethacin ingestion plus ATP administration [0.027 (0.020-0.046)] was significantly lower than the lactulose/rhamnose ratio in the placebo condition (P<0.01). CONCLUSIONS Topical ATP administration into the small intestine during short-term challenge of the NSAID indomethacin attenuates the NSAID-induced increase in small intestinal permeability in healthy humans.
Collapse
Affiliation(s)
- Martijn J Bours
- Department of Epidemiology, Maastricht University, Nutrition and Toxicology Research Institute Maastricht, Maastricht, The Netherlands.
| | | | | | | | | |
Collapse
|
8
|
Richter Y, Fischer B. Nucleotides and inorganic phosphates as potential antioxidants. J Biol Inorg Chem 2006; 11:1063-74. [PMID: 16896806 DOI: 10.1007/s00775-006-0143-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2006] [Accepted: 06/30/2006] [Indexed: 10/24/2022]
Abstract
Highly reactive OH radicals, formed in an iron-ion catalyzed Fenton reaction, are implicated in many pathological conditions. The quest for Fenton reaction inhibitors, either radical scavenger or metal-ion chelator antioxidants, spans the previous decades. Purine nucleotides were previously studied as natural modulators of the Fenton reaction; however, the modulatory role of purine nucleotides remained in dispute. Here, we have resolved this long-standing dispute and demonstrated a concentration-dependent biphasic modulation of the Fenton reaction by nucleotides. By electron spin resonance measurements with 0.1 mM Fe(II), we observed an increase of *OH production at low purine nucleotide concentrations (up to 0.15 mM), while at higher nucleotide concentrations, an exponential decay of *OH concentration was observed. We found that the phosphate moiety, not the nucleoside, determines the pro/antioxidant properties of a nucleotide, suggesting a chelation-based modulation. Furthermore, the biphasic modulation mode is probably due to diverse nucleotide-Fe(II) complexes formed in a concentration-dependent manner. At ATP concentrations much greater than Fe(II) concentrations, multiligand chelates are formed which inhibit the Fenton reaction owing to a full Fe(II) coordination sphere. In addition to natural nucleotides, we investigated a series of base- or phosphate-modified nucleotides, dinucleotides, and inorganic phosphates, as potential biocompatible antioxidants. Ap5A, inorganic thiophosphate and ATP-gamma-S proved highly potent antioxidants with IC50 values of 40, 30, and 10 microM, respectively. ATP-gamma-S proved 100 and 20 times more active than ATP and the potent antioxidant Trolox, respectively. In the presence of 30 microM ATP-gamma-S no *OH was detected after 5 min in the Fenton reaction mixture. The most potent antioxidants identified inhibit the Fenton reaction by forming full coordination sphere chelates.
Collapse
Affiliation(s)
- Yael Richter
- Department of Chemistry, Gonda-Goldschmied Medical Research Center, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | | |
Collapse
|
9
|
Bours MJL, Swennen ELR, Di Virgilio F, Cronstein BN, Dagnelie PC. Adenosine 5'-triphosphate and adenosine as endogenous signaling molecules in immunity and inflammation. Pharmacol Ther 2006; 112:358-404. [PMID: 16784779 DOI: 10.1016/j.pharmthera.2005.04.013] [Citation(s) in RCA: 776] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2005] [Accepted: 04/20/2005] [Indexed: 02/07/2023]
Abstract
Human health is under constant threat of a wide variety of dangers, both self and nonself. The immune system is occupied with protecting the host against such dangers in order to preserve human health. For that purpose, the immune system is equipped with a diverse array of both cellular and non-cellular effectors that are in continuous communication with each other. The naturally occurring nucleotide adenosine 5'-triphosphate (ATP) and its metabolite adenosine (Ado) probably constitute an intrinsic part of this extensive immunological network through purinergic signaling by their cognate receptors, which are widely expressed throughout the body. This review provides a thorough overview of the effects of ATP and Ado on major immune cell types. The overwhelming evidence indicates that ATP and Ado are important endogenous signaling molecules in immunity and inflammation. Although the role of ATP and Ado during the course of inflammatory and immune responses in vivo appears to be extremely complex, we propose that their immunological role is both interdependent and multifaceted, meaning that the nature of their effects may shift from immunostimulatory to immunoregulatory or vice versa depending on extracellular concentrations as well as on expression patterns of purinergic receptors and ecto-enzymes. Purinergic signaling thus contributes to the fine-tuning of inflammatory and immune responses in such a way that the danger to the host is eliminated efficiently with minimal damage to healthy tissues.
Collapse
Affiliation(s)
- M J L Bours
- Maastricht University, Department of Epidemiology, Nutrition and Toxicology Research Institute Maastricht, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | | | | | | | | |
Collapse
|
10
|
Invited Lectures : Overviews Purinergic signalling: past, present and future. Purinergic Signal 2006; 2:1-324. [PMID: 18404494 PMCID: PMC2096525 DOI: 10.1007/s11302-006-9006-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2006] [Indexed: 12/11/2022] Open
|