1
|
Lungu PF, Lungu CM, Ciobica A, Balmus IM, Vitalaru R, Mavroudis I, Dobrin R, Cimpeanu M, Gurzu IL. The Effect of Antipsychotics on Cognition in Schizophrenia-A Current Narrative Review. Brain Sci 2024; 14:359. [PMID: 38672011 PMCID: PMC11047912 DOI: 10.3390/brainsci14040359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/14/2024] [Accepted: 03/31/2024] [Indexed: 04/28/2024] Open
Abstract
The majority of schizophrenia-affected individuals display deficiencies in multiple cognitive domains such as attention, working memory, long-term memory, and learning, deficiencies that are stable throughout the disease. The purpose of this narrative review was to examine the effect of antipsychotics on several cognitive domains affected by schizophrenia. Methods: We searched MEDLINE, Elsevier, Scopus, and DOAJ databases for randomized controlled trials and other studies investigating the effects of typical and atypical antipsychotics on cognition in patients with schizophrenia in studies conducted in the last decade. Results: The majority of studies included in this review showed that antipsychotics (especially SGAs) have positive effects on both cognition and general psychopathology of schizophrenia. We mention that treatment with antipsychotic substances represents an ongoing effort of the researchers, who are constantly searching for the best approach to meet the mental health needs of schizophrenia patients. Conclusions: Even with those positive results, it should be noted that more studies are needed in order to fully observe the various effects of certain antipsychotic substances on cognition.
Collapse
Affiliation(s)
- Petru Fabian Lungu
- Faculty of Biology, Biology Department, “Alexandru Ioan Cuza” University, 700506 Iasi, Romania; (P.F.L.); (M.C.)
| | - Corina Miruna Lungu
- Faculty of Psychology and Educational Sciences, Psychology Department, “Alexandru Ioan Cuza” University, 700506 Iasi, Romania
| | - Alin Ciobica
- Faculty of Biology, Biology Department, “Alexandru Ioan Cuza” University, 700506 Iasi, Romania; (P.F.L.); (M.C.)
- Center of Biomedical Research, Romanian Academy, Iasi Branch, Teodor Codrescu 2, 700481 Iasi, Romania
- Academy of Romanian Scientists, 3 Ilfov, 050044 Bucharest, Romania
- Preclinical Department, Apollonia University, Strada Păcurari 11, 700511 Iasi, Romania
| | - Ioana Miruna Balmus
- Institute of Interdisciplinary Research, Department of Exact Sciences and Natural Sciences, “Alexandru Ioan Cuza” University, 700506 Iasi, Romania;
- CENEMED Platform for Interdisciplinary Research, “Grigore T. Popa” University of Medicine and Pharmacy of Iasi, 16th Universitatii Street, 700115 Iasi, Romania
| | - Raluca Vitalaru
- Institute of Psychiatry “Socola”, Iasi Str. Bucium 36, 700282 Iasi, Romania (R.D.)
| | - Ioannis Mavroudis
- Department of Neuroscience, Leeds Teaching Hospitals, NHS Trust, Leeds LS2 9JT, UK
| | - Romeo Dobrin
- Institute of Psychiatry “Socola”, Iasi Str. Bucium 36, 700282 Iasi, Romania (R.D.)
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania;
| | - Mirela Cimpeanu
- Faculty of Biology, Biology Department, “Alexandru Ioan Cuza” University, 700506 Iasi, Romania; (P.F.L.); (M.C.)
| | - Irina Luciana Gurzu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania;
| |
Collapse
|
2
|
Daliri DB, Afaya A, Laari TT, Fredrick A, Ankamah AG, Annan S, Abagye N. Five-year trend of antipsychotic prescription practices in a district Hospital in Ghana: A retrospective study. Neuropsychopharmacol Rep 2023; 43:434-439. [PMID: 37574802 PMCID: PMC10496068 DOI: 10.1002/npr2.12372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/15/2023] Open
Abstract
INTRODUCTION Antipsychotics are the mainstay treatment for psychotic conditions. Their prescription, however, should come with some caution since the consequences of their side effects can be dire for the patient receiving the prescription. Because of inadequate experts in low-middle-income countries, non-experts are trained through the Mental Health Gap Action Program (MHGAP) to reduce the treatment gap. This retrospective study analyzed the trend of antipsychotic prescriptions in a district hospital where mental health services are delivered by non-experts. METHODOLOGY This was a retrospective descriptive study that gathered data between 2015 and 2019 from the electronic database of the hospital. Statistical analysis was conducted using SPSS version 20. We reported the descriptive statistics of our findings in the form of frequencies and percentages. RESULTS There was a year-on-year increase in antipsychotic prescriptions over the study period. Starting with 48.1% in 2015 to 56.4% in 2019. The main condition for which antipsychotics were prescribed was psychosis (58.6%), followed by substance use disorder (SUD) (26%). Patients with age ≥ 50 received the most prescription of antipsychotics. Starting from 2015, there was a high percentage of typical antipsychotic prescriptions (90.14%) with atypical antipsychotics being 9.86% and by 2019 atypical antipsychotic prescriptions had shot up to 74.8%. Polypharmacy prescription rate was 8.1% over the study period. CONCLUSION Antipsychotics are essential in the treatment of psychosis and other mental health conditions. Prescribers need to know more about these drugs to prescribe them appropriately and to minimize the likelihood of side effects among patients who use these drugs.
Collapse
Affiliation(s)
- Dennis Bomansang Daliri
- Department of Global Health, School of Public HealthUniversity for Development studiesTamaleGhana
- Presbyterian Psychiatric HospitalBolgatangaGhana
| | - Agani Afaya
- Department of Nursing, School of Nursing and MidwiferyUniversity of Health and Allied sciencesHoGhana
- Mo‐Im Kim Nursing Research Institute, College of Nursing, Yonsei UniversitySeoulSouth Korea
| | | | | | | | | | - Nancy Abagye
- Department of Midwifery, School of Nursing and MidwiferyUniversity of GhanaAccraGhana
| |
Collapse
|
3
|
Voss JH, Crüsemann M, Bartling CR, Kehraus S, Inoue A, König GM, Strømgaard K, Müller CE. Structure-affinity and structure-residence time relationships of macrocyclic Gα q protein inhibitors. iScience 2023; 26:106492. [PMID: 37091255 PMCID: PMC10119753 DOI: 10.1016/j.isci.2023.106492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/02/2023] [Accepted: 03/21/2023] [Indexed: 04/25/2023] Open
Abstract
The macrocyclic depsipeptides YM-254890 (YM) and FR900359 (FR) are potent inhibitors of Gαq/11 proteins. They are important pharmacological tools and have potential as therapeutic drugs. The hydrogenated, tritium-labeled YM and FR derivatives display largely different residence times despite similar structures. In the present study we established a competition-association binding assay to determine the dissociation kinetics of unlabeled Gq protein inhibitors. Structure-affinity and structure-residence time relationships were analyzed. Small structural modifications had a large impact on residence time. YM and FR exhibited 4- to 10-fold higher residence times than their hydrogenated derivatives. While FR showed pseudo-irreversible binding, YM displayed much faster dissociation from its target. The isopropyl anchor present in FR and some derivatives was essential for slow dissociation. These data provide a basis for future drug design toward modulating residence times of macrocyclic Gq protein inhibitors, which has been recognized as a crucial determinant for therapeutic outcome.
Collapse
Affiliation(s)
- Jan H. Voss
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Max Crüsemann
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Christian R.O. Bartling
- Department of Drug Design and Pharmacology, Center for Biopharmaceuticals, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Stefan Kehraus
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Asuka Inoue
- Tohoku University, Graduate School of Pharmaceutical Sciences, Sendai, Miyagi 980-8578, Japan
| | - Gabriele M. König
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, Center for Biopharmaceuticals, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Christa E. Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
- Corresponding author
| |
Collapse
|
4
|
Xu Q, Mao Z, Yun Y. Adenosine A 1 receptor agonism protection mechanism in intestinal ischemia/reperfusion injury via activation of PI3K/Akt signaling. Exp Ther Med 2022; 25:41. [PMID: 36569432 PMCID: PMC9764055 DOI: 10.3892/etm.2022.11740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
Intestinal ischemia/reperfusion (I/R) injury is a common clinical problem with a high mortality rate, resulting from loss of blood flow to an intestinal segment. Adenosine serves a protective role in intestinal I/R injury; however, its potential mechanism is not completely understood. The present study aimed to investigate the protective effects of adenosine A1 receptor (A1R) agonists CPA and LUF6941 and whether their mechanisms are associated with the PI3K/Akt signaling pathway. To simulate intestinal I/R injury, a cell oxygen-glucose deprivation/reoxygenation (OGD/R) model was established and the human colon cancer cell line (Caco-2) was incubated with A1R agonists before OGD/R treatment. The viability of Caco-2 cells was detected by PI and Cell Counting Kit-8 assay, apoptosis was detected using flow cytometry and western blotting was used to analyze protein expression levels of PI3K, Akt and p53 in Caco-2 cells. A1R agonist pretreatment protected Caco-2 cells against OGD/R-induced cell damage and activated PI3K/Akt signaling. Additionally, apoptosis was inhibited by downregulating phosphorylation of p53 protein, as evidenced by increased cell viability. These findings suggested that A1R agonists decreased OGD/R damage in Caco-2 cells, which may be due to their anti-apoptotic effects and activation of the PI3K/Akt/p53 signal pathway.
Collapse
Affiliation(s)
- Qian Xu
- Department of Pharmacy, Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, Jiangsu 223800, P.R. China,Department of Gastroenterology, Affiliated Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Zun Mao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, P.R. China
| | - Yi Yun
- Department of Pharmacy, Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, Jiangsu 223800, P.R. China,Correspondence to: Dr Yi Yun, Department of Pharmacy, Affiliated Suqian First People's Hospital of Nanjing Medical University, 120 Suzhilu Street, Sucheng, Suqian, Jiangsu 223800, P.R. China
| |
Collapse
|
5
|
Zhu MH, Liu ZJ, Hu QY, Yang JY, Jin Y, Zhu N, Huang Y, Shi DH, Liu MJ, Tan HY, Zhao L, Lv QY, Yi ZH, Wu FC, Li ZZ. Amisulpride augmentation therapy improves cognitive performance and psychopathology in clozapine-resistant treatment-refractory schizophrenia: a 12-week randomized, double-blind, placebo-controlled trial. Mil Med Res 2022; 9:59. [PMID: 36253804 PMCID: PMC9578180 DOI: 10.1186/s40779-022-00420-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 09/19/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Although clozapine is an effective option for treatment-resistant schizophrenia (TRS), there are still 1/3 to 1/2 of TRS patients who do not respond to clozapine. The main purpose of this randomized, double-blind, placebo-controlled trial was to explore the amisulpride augmentation efficacy on the psychopathological symptoms and cognitive function of clozapine-resistant treatment-refractory schizophrenia (CTRS) patients. METHODS A total of 80 patients were recruited and randomly assigned to receive initial clozapine plus amisulpride (amisulpride group) or clozapine plus placebo (placebo group). Positive and Negative Syndrome Scale (PANSS), Scale for the Assessment of Negative Symptoms (SANS), Clinical Global Impression (CGI) scale scores, Repeatable Battery for the Assessment of Neuropsychological Status (RBANS), Treatment Emergent Symptom Scale (TESS), laboratory measurements, and electrocardiograms (ECG) were performed at baseline, at week 6, and week 12. RESULTS Compared with the placebo group, amisulpride group had a lower PANSS total score, positive subscore, and general psychopathology subscore at week 6 and week 12 (PBonferroni < 0.01). Furthermore, compared with the placebo group, the amisulpride group showed an improved RBANS language score at week 12 (PBonferroni < 0.001). Amisulpride group had a higher treatment response rate (P = 0.04), lower scores of CGI severity and CGI efficacy at week 6 and week 12 than placebo group (PBonferroni < 0.05). There were no differences between the groups in body mass index (BMI), corrected QT (QTc) intervals, and laboratory measurements. This study demonstrates that amisulpride augmentation therapy can safely improve the psychiatric symptoms and cognitive performance of CTRS patients. CONCLUSION This study indicates that amisulpride augmentation therapy has important clinical significance for treating CTRS to improve clinical symptoms and cognitive function with tolerability and safety. Trial registration Clinicaltrials.gov identifier- NCT03652974. Registered August 31, 2018, https://clinicaltrials.gov/ct2/show/NCT03652974.
Collapse
Affiliation(s)
- Ming-Huan Zhu
- Clinical Research Center for Mental Disorders, School of Medicine, Shanghai Pudong New Area Mental Health Center, Tongji University, Shanghai, 200124, China
| | - Zhen-Jing Liu
- Qingdao Mental Health Center, Qingdao, 266034, Shandong, China
| | - Qiong-Yue Hu
- Qingdao Mental Health Center, Qingdao, 266034, Shandong, China
| | - Jia-Yu Yang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Ying Jin
- Clinical Research Center for Mental Disorders, School of Medicine, Shanghai Pudong New Area Mental Health Center, Tongji University, Shanghai, 200124, China
| | - Na Zhu
- Clinical Research Center for Mental Disorders, School of Medicine, Shanghai Pudong New Area Mental Health Center, Tongji University, Shanghai, 200124, China
| | - Ying Huang
- Clinical Research Center for Mental Disorders, School of Medicine, Shanghai Pudong New Area Mental Health Center, Tongji University, Shanghai, 200124, China
| | - Dian-Hong Shi
- Clinical Research Center for Mental Disorders, School of Medicine, Shanghai Pudong New Area Mental Health Center, Tongji University, Shanghai, 200124, China
| | - Min-Jia Liu
- Clinical Research Center for Mental Disorders, School of Medicine, Shanghai Pudong New Area Mental Health Center, Tongji University, Shanghai, 200124, China
| | - Hong-Yang Tan
- Clinical Research Center for Mental Disorders, School of Medicine, Shanghai Pudong New Area Mental Health Center, Tongji University, Shanghai, 200124, China
| | - Lei Zhao
- Qingdao Mental Health Center, Qingdao, 266034, Shandong, China
| | - Qin-Yu Lv
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Zheng-Hui Yi
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Feng-Chun Wu
- Department of Psychiatry, the Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, 510370, China. .,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China.
| | - Ze-Zhi Li
- Department of Psychiatry, the Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, 510370, China. .,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China.
| |
Collapse
|
6
|
Al Abadey A, Connor B, Flamme ACL, Robichon K. Clozapine reduces chemokine-mediated migration of lymphocytes by targeting NF-κB and AKT phosphorylation. Cell Signal 2022; 99:110449. [PMID: 36031090 DOI: 10.1016/j.cellsig.2022.110449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/11/2022] [Accepted: 08/22/2022] [Indexed: 11/27/2022]
Abstract
Multiple sclerosis is a disease characterised by demyelination of axons in the central nervous system. The atypical antipsychotic drug clozapine has been shown to attenuate disease severity in experimental autoimmune encephalomyelitis (EAE), a mouse model that is useful for the study of multiple sclerosis. However, the mechanism of action by which clozapine reduces disease in EAE is poorly understood. To better understand how clozapine exerts its protective effects, we investigated the underlying signalling pathways by which clozapine may reduce immune cell migration by evaluating chemokine and dopamine receptor-associated signalling pathways. We found that clozapine inhibits migration of immune cells by reducing chemokine production in microglia cells by targeting NF-κB phosphorylation and promoting an anti-inflammatory milieu. Furthermore, clozapine directly targets immune cell migration by changing Ca2+ levels within immune cells and reduces the phosphorylation of signalling protein AKT. Linking these pathways to the antagonising effect of clozapine on dopamine and serotonin receptors, we provide insight into how clozapine alters immune cells migration by directly targeting the underlying migration-associated pathways.
Collapse
Affiliation(s)
- Afnan Al Abadey
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand; Centre for Biodiscovery Wellington, Victoria University of Wellington, Wellington, New Zealand
| | - Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Anne Camille La Flamme
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand; Centre for Biodiscovery Wellington, Victoria University of Wellington, Wellington, New Zealand; Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Katharina Robichon
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand; Centre for Biodiscovery Wellington, Victoria University of Wellington, Wellington, New Zealand.
| |
Collapse
|
7
|
Ahmad K, Rizzi A, Capelli R, Mandelli D, Lyu W, Carloni P. Enhanced-Sampling Simulations for the Estimation of Ligand Binding Kinetics: Current Status and Perspective. Front Mol Biosci 2022; 9:899805. [PMID: 35755817 PMCID: PMC9216551 DOI: 10.3389/fmolb.2022.899805] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022] Open
Abstract
The dissociation rate (k off) associated with ligand unbinding events from proteins is a parameter of fundamental importance in drug design. Here we review recent major advancements in molecular simulation methodologies for the prediction of k off. Next, we discuss the impact of the potential energy function models on the accuracy of calculated k off values. Finally, we provide a perspective from high-performance computing and machine learning which might help improve such predictions.
Collapse
Affiliation(s)
- Katya Ahmad
- Computational Biomedicine (IAS-5/INM-9), Forschungszentrum Jülich, Jülich, Germany
| | - Andrea Rizzi
- Computational Biomedicine (IAS-5/INM-9), Forschungszentrum Jülich, Jülich, Germany
- Atomistic Simulations, Istituto Italiano di Tecnologia, Genova, Italy
| | - Riccardo Capelli
- Department of Applied Science and Technology (DISAT), Politecnico di Torino, Torino, Italy
| | - Davide Mandelli
- Computational Biomedicine (IAS-5/INM-9), Forschungszentrum Jülich, Jülich, Germany
| | - Wenping Lyu
- Warshel Institute for Computational Biology, School of Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, China
| | - Paolo Carloni
- Computational Biomedicine (IAS-5/INM-9), Forschungszentrum Jülich, Jülich, Germany
- Molecular Neuroscience and Neuroimaging (INM-11), Forschungszentrum Jülich, Jülich, Germany
| |
Collapse
|
8
|
Distinct In Vitro Binding Profile of the Somatostatin Receptor Subtype 2 Antagonist [ 177Lu]Lu-OPS201 Compared to the Agonist [ 177Lu]Lu-DOTA-TATE. Pharmaceuticals (Basel) 2021; 14:ph14121265. [PMID: 34959665 PMCID: PMC8706879 DOI: 10.3390/ph14121265] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 01/14/2023] Open
Abstract
Treatment of neuroendocrine tumours with the radiolabelled somatostatin receptor subtype 2 (SST2) peptide agonist [177Lu]Lu-DOTA-TATE is effective and well-established. Recent studies suggest improved therapeutic efficacy using the SST2 peptide antagonist [177Lu]Lu-OPS201. However, little is known about the cellular mechanisms that lead to the observed differences. In the present in vitro study, we compared kinetic binding, saturation binding, competition binding, cellular uptake and release of [177Lu]Lu-OPS201 versus [177Lu]Lu-DOTA-TATE using HEK cells stably transfected with the human SST2. While [177Lu]Lu-OPS201 and [177Lu]Lu-DOTA-TATE exhibited comparable affinity (KD, 0.15 ± 0.003 and 0.08 ± 0.02 nM, respectively), [177Lu]Lu-OPS201 recognized four times more binding sites than [177Lu]Lu-DOTA-TATE. Competition assays demonstrated that a high concentration of the agonist displaced only 30% of [177Lu]Lu-OPS201 bound to HEK-SST2 cell membranes; an indication that the antagonist binds to additional sites that are not recognized by the agonist. [177Lu]Lu-OPS201 showed faster association and slower dissociation than [177Lu]Lu-DOTA-TATE. Whereas most of [177Lu]Lu-OPS201 remained at the cell surface, [177Lu]Lu-DOTA-TATE was almost completely internalised inside the cell. The present data identified distinct differences between [177Lu]Lu-OPS201 and [177Lu]Lu-DOTA-TATE regarding the recognition of receptor binding sites (higher for [177Lu]Lu-OPS201) and their kinetics (faster association and slower dissociation of [177Lu]Lu-OPS201) that explain, to a great extent, the improved therapeutic efficacy of [177Lu]Lu-OPS201 compared to [177Lu]Lu-DOTA-TATE.
Collapse
|
9
|
G protein signaling-biased mu opioid receptor agonists that produce sustained G protein activation are noncompetitive agonists. Proc Natl Acad Sci U S A 2021; 118:2102178118. [PMID: 34819362 DOI: 10.1073/pnas.2102178118] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 12/15/2022] Open
Abstract
The ability of a ligand to preferentially promote engagement of one signaling pathway over another downstream of GPCR activation has been referred to as signaling bias, functional selectivity, and biased agonism. The presentation of ligand bias reflects selectivity between active states of the receptor, which may result in the display of preferential engagement with one signaling pathway over another. In this study, we provide evidence that the G protein-biased mu opioid receptor (MOR) agonists SR-17018 and SR-14968 stabilize the MOR in a wash-resistant yet antagonist-reversible G protein-signaling state. Furthermore, we demonstrate that these structurally related biased agonists are noncompetitive for radiolabeled MOR antagonist binding, and while they stimulate G protein signaling in mouse brains, partial agonists of this class do not compete with full agonist activation. Importantly, opioid antagonists can readily reverse their effects in vivo. Given that chronic treatment with SR-17018 does not lead to tolerance in several mouse pain models, this feature may be desirable for the development of long-lasting opioid analgesics that remain sensitive to antagonist reversal of respiratory suppression.
Collapse
|
10
|
Seeman MV. History of the dopamine hypothesis of antipsychotic action. World J Psychiatry 2021; 11:355-364. [PMID: 34327128 PMCID: PMC8311512 DOI: 10.5498/wjp.v11.i7.355] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/22/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
The dopamine hypothesis of how antipsychotic drugs exert their beneficial effect in psychotic illness has an interesting history that dates back to 1950. This hypothesis is not to be confused with the dopamine hypothesis of schizophrenia; the aim of the latter is to explain the etiology of schizophrenia. The present review does not deal with schizophrenia but, rather, with the historical development of our current understanding of the dopamine-associated actions of the drugs that reduce the symptoms of psychosis. This historical review begins with the serendipitous discovery of chlorpromazine, a drug synthesized around a chemical core that initially served to produce man-made dyes. This molecular core subsequently contributed to the chemistry of antihistamines. It was with the aim of producing a superior antihistamine that chlorpromazine was synthesized; instead, it revolutionized the treatment of psychosis. The first hypothesis of how this drug worked was that it induced hypothermia, a cooling of the body that led to a tranquilization of the mind. The new, at the time, discoveries of the presence of chemical transmitters in the brain soon steered investigations away from a temperature-related hypothesis toward questioning how this drug, and other drugs with similar properties and effects, modulated endogenous neurotransmission. As a result, over the years, researchers from around the world have begun to progressively learn what antipsychotic drugs do in the brain.
Collapse
Affiliation(s)
- Mary V Seeman
- Department of Psychiatry, University of Toronto, Toronto M5P 3L6, Ontario, Canada
| |
Collapse
|
11
|
New small molecule fluorescent probes for G protein-coupled receptors: valuable tools for drug discovery. Future Med Chem 2020; 13:63-90. [PMID: 33319586 DOI: 10.4155/fmc-2019-0327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are essential signaling proteins and tractable therapeutic targets. To develop new drug candidates, GPCR drug discovery programs require versatile, sensitive pharmacological tools for ligand binding and compound screening. With the availability of new imaging modalities and proximity-based ligand binding technologies, fluorescent ligands offer many advantages and are increasingly being used, yet labeling small molecules remains considerably more challenging relative to peptides. Focusing on recent fluorescent small molecule studies for family A GPCRs, this review addresses some of the key challenges, synthesis approaches and structure-activity relationship considerations, and discusses advantages of using high-resolution GPCR structures to inform conjugation strategies. While no single approach guarantees successful labeling without loss of affinity or selectivity, the choice of fluorophore, linker type and site of attachment have proved to be critical factors that can significantly affect their utility in drug discovery programs, and as discussed, can sometimes lead to very unexpected results.
Collapse
|
12
|
Schuetz DA, Richter L, Martini R, Ecker GF. A structure-kinetic relationship study using matched molecular pair analysis. RSC Med Chem 2020; 11:1285-1294. [PMID: 34085042 PMCID: PMC8126976 DOI: 10.1039/d0md00178c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The lifetime of a binary drug–target complex is increasingly acknowledged as an important parameter for drug efficacy and safety. With a better understanding of binding kinetics and better knowledge about kinetic parameter optimization, intentionally induced prolongation of the drug–target residence time through structural changes of the ligand could become feasible. In this study we assembled datasets from 21 publications and the K4DD (Kinetic for Drug Discovery) database to conduct large scale data analysis. This resulted in 3812 small molecules annotated to 78 different targets from five protein classes (GPCRs: 273, kinases: 3238, other enzymes: 240, HSPs: 160, ion channels: 45). Performing matched molecular pair (MMP) analysis to further investigate the structure–kinetic relationship (SKR) in this data collection allowed us to identify a fundamental contribution of a ligand's polarity to its association rate, and in selected cases, also to its dissociation rate. However, we furthermore observed that the destabilization of the transition state introduced by increased polarity is often accompanied by simultaneous destabilization of the ground state resulting in an unaffected or even worsened residence time. Supported by a set of case studies, we provide concepts on how to alter ligands in ways to trigger on-rates, off-rates, or both. A large-scale study employing matched molecular pair (MMP) analysis to uncover the contribution of a compound's polarity to its association and dissociation rates.![]()
Collapse
Affiliation(s)
- Doris A Schuetz
- Department of Pharmaceutical Chemistry, University of Vienna UZA 2, Althanstrasse 14 1090 Vienna Austria
| | - Lars Richter
- Department of Pharmaceutical Chemistry, University of Vienna UZA 2, Althanstrasse 14 1090 Vienna Austria
| | - Riccardo Martini
- Department of Pharmaceutical Chemistry, University of Vienna UZA 2, Althanstrasse 14 1090 Vienna Austria
| | - Gerhard F Ecker
- Department of Pharmaceutical Chemistry, University of Vienna UZA 2, Althanstrasse 14 1090 Vienna Austria
| |
Collapse
|
13
|
Hall R, Dixon T, Dickson A. On Calculating Free Energy Differences Using Ensembles of Transition Paths. Front Mol Biosci 2020; 7:106. [PMID: 32582764 PMCID: PMC7291376 DOI: 10.3389/fmolb.2020.00106] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/06/2020] [Indexed: 12/30/2022] Open
Abstract
The free energy of a process is the fundamental quantity that determines its spontaneity or propensity at a given temperature. In particular, the binding free energy of a drug candidate to its biomolecular target is used as an objective quantity in drug design. Recently, binding kinetics—rates of association (kon) and dissociation (koff)—have also demonstrated utility for their ability to predict efficacy and in some cases have been shown to be more predictive than the binding free energy alone. Some methods exist to calculate binding kinetics from molecular simulations, although these are typically more difficult to calculate than the binding affinity as they depend on details of the transition path ensemble. Assessing these rate constants can be difficult, due to uncertainty in the definition of the bound and unbound states, large error bars and the lack of experimental data. As an additional consistency check, rate constants from simulation can be used to calculate free energies (using the log of their ratio) which can then be compared to free energies obtained experimentally or using alchemical free energy perturbation. However, in this calculation it is not straightforward to account for common, practical details such as the finite simulation volume or the particular definition of the “bound” and “unbound” states. Here we derive a set of correction terms that can be applied to calculations of binding free energies using full reactive trajectories. We apply these correction terms to revisit the calculation of binding free energies from rate constants for a host-guest system that was part of a blind prediction challenge, where significant deviations were observed between free energies calculated with rate ratios and those calculated from alchemical perturbation. The correction terms combine to significantly decrease the error with respect to computational benchmarks, from 3.4 to 0.76 kcal/mol. Although these terms were derived with weighted ensemble simulations in mind, some of the correction terms are generally applicable to free energies calculated using physical pathways via methods such as Markov state modeling, metadynamics, milestoning, or umbrella sampling.
Collapse
Affiliation(s)
- Robert Hall
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI, United States
| | - Tom Dixon
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI, United States.,Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Alex Dickson
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI, United States.,Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
14
|
Limongelli V. Ligand binding free energy and kinetics calculation in 2020. WILEY INTERDISCIPLINARY REVIEWS-COMPUTATIONAL MOLECULAR SCIENCE 2020. [DOI: 10.1002/wcms.1455] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Vittorio Limongelli
- Faculty of Biomedical Sciences, Institute of Computational Science – Center for Computational Medicine in Cardiology Università della Svizzera italiana (USI) Lugano Switzerland
- Department of Pharmacy University of Naples “Federico II” Naples Italy
| |
Collapse
|
15
|
Borisov DV, Veselovsky AV. [Ligand-receptor binding kinetics in drug design]. BIOMEDITSINSKAIA KHIMIIA 2020; 66:42-53. [PMID: 32116225 DOI: 10.18097/pbmc20206601042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Traditionally, the thermodynamic values of affinity are considered as the main criterion for the development of new drugs. Usually, these values for drugs are measured <i>in vitro</i> at steady concentrations of the receptor and ligand, which are differed from <i>in vivo</i> environment. Recent studies have shown that the kinetics of the process of drug binding to its receptor make significant contribution in the drug effectiveness. This has increased attention in characterizing and predicting the rate constants of association and dissociation of the receptor ligand at the stage of preclinical studies of drug candidates. A drug with a long residence time can determine ligand-receptor selectivity (kinetic selectivity), maintain pharmacological activity of the drug at its low concentration in vivo. The paper discusses the theoretical basis of protein-ligand binding, molecular determinants that control the kinetics of the drug-receptor binding. Understanding the molecular features underlying the kinetics of receptor-ligand binding will contribute to the rational design of drugs with desired properties.
Collapse
Affiliation(s)
- D V Borisov
- Institute of Biomedical Chemistry, Moscow, Russia
| | | |
Collapse
|
16
|
Long residence time adenosine A1 receptor agonists produce sustained wash-resistant antilipolytic effect in rat adipocytes. Biochem Pharmacol 2019; 164:45-52. [DOI: 10.1016/j.bcp.2019.03.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 03/20/2019] [Indexed: 02/06/2023]
|
17
|
Chang H, Wei Y, Chen Y, Du L, Cong H, Zhang X, Geng X, Yin L. The antipsychotic-like effects of clozapine in C57BL/6 mice exposed to cuprizone: Decreased glial activation. Behav Brain Res 2019; 364:157-161. [DOI: 10.1016/j.bbr.2019.02.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 12/14/2022]
|
18
|
Sykes DA, Stoddart LA, Kilpatrick LE, Hill SJ. Binding kinetics of ligands acting at GPCRs. Mol Cell Endocrinol 2019; 485:9-19. [PMID: 30738950 PMCID: PMC6406023 DOI: 10.1016/j.mce.2019.01.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/19/2019] [Accepted: 01/19/2019] [Indexed: 12/31/2022]
Abstract
The influence of drug-receptor binding kinetics has often been overlooked during the development of new therapeutics that target G protein-coupled receptors (GPCRs). Over the last decade there has been a growing understanding that an in-depth knowledge of binding kinetics at GPCRs is required to successfully target this class of proteins. Ligand binding to a GPCR is often not a simple single step process with ligand freely diffusing in solution. This review will discuss the experiments and equations that are commonly used to measure binding kinetics and how factors such as allosteric regulation, rebinding and ligand interaction with the plasma membrane may influence these measurements. We will then consider the molecular characteristics of a ligand and if these can be linked to association and dissociation rates.
Collapse
Affiliation(s)
- David A Sykes
- Cell Signalling and Pharmacology Research Group, Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Leigh A Stoddart
- Cell Signalling and Pharmacology Research Group, Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Laura E Kilpatrick
- Cell Signalling and Pharmacology Research Group, Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Stephen J Hill
- Cell Signalling and Pharmacology Research Group, Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK.
| |
Collapse
|
19
|
Waszak PM, Zagożdżon P, Pierucka M, Kubanek A. Antipsychotic Medication Prescribing Trends in a Pediatric Population in Northern Poland 2008-2012. J Child Adolesc Psychopharmacol 2018; 28:631-636. [PMID: 30048153 DOI: 10.1089/cap.2017.0154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVES In recent years antipsychotics have been increasingly used in the population of children and adolescents. The objective of this article is to profile the use of antipsychotic medications in a pediatric population in Poland based on data from the Pomeranian region (Northern Poland). METHODS The study was based on National Health Fund data about prescription drug reimbursement between 2008 and 2012. A patient database was created using ID to analyze specific patterns and information about individuals using antipsychotics. Percentage population indicators were calculated using official demographic reports. RESULTS An increased number of overall prescriptions was observed since 2008, with a subsequent decline to its lowest number in 2012. The population with at least one antipsychotic-filled prescription per year has grown in the same time. The annual rate has increased from 0.26% to 0.31% of the general population between 0 and 17 years of age in the Pomeranian region. We observed an increasing number of females and the 0- to 4-year-old receiving antipsychotic prescriptions. The population share of prescribed first-generation antipsychotics exceeded the share of second-generation antipsychotics (SGAs). The use of SGAs increased from 38% to 44% of all prescriptions, during the observation period. The most frequently prescribed drugs were Risperidone (26.7%) and Chlorprothixene (21.7%). CONCLUSIONS The study revealed that the prescribing patterns of antipsychotics increased in the Polish pediatric population and concerned more patients from the youngest group. The high prevalence of such early exposure to antipsychotics should focus more attention on drug safety in this population.
Collapse
Affiliation(s)
- Przemysław M Waszak
- 1 Department of Hygiene and Epidemiology, Medical University of Gdansk , Gdansk, Poland .,2 Department of Developmental Psychiatry, Psychotic and Geriatric Disorders, Medical University of Gdansk , Gdansk, Poland
| | - Paweł Zagożdżon
- 1 Department of Hygiene and Epidemiology, Medical University of Gdansk , Gdansk, Poland
| | - Magdalena Pierucka
- 1 Department of Hygiene and Epidemiology, Medical University of Gdansk , Gdansk, Poland
| | - Alicja Kubanek
- 3 Department of Occupational, Metabolic, and Internal Diseases, Medical University of Gdansk , Gdansk, Poland
| |
Collapse
|
20
|
de Witte WEA, Versfelt JW, Kuzikov M, Rolland S, Georgi V, Gribbon P, Gul S, Huntjens D, van der Graaf PH, Danhof M, Fernández-Montalván A, Witt G, de Lange ECM. In vitro and in silico analysis of the effects of D 2 receptor antagonist target binding kinetics on the cellular response to fluctuating dopamine concentrations. Br J Pharmacol 2018; 175:4121-4136. [PMID: 30051456 PMCID: PMC6177617 DOI: 10.1111/bph.14456] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 06/17/2018] [Accepted: 06/25/2018] [Indexed: 12/27/2022] Open
Abstract
Background and Purpose Target binding kinetics influence the time course of the drug effect (pharmacodynamics) both (i) directly, by affecting the time course of target occupancy, driven by the pharmacokinetics of the drug, competition with endogenous ligands and target turnover, and (ii) indirectly, by affecting signal transduction and homeostatic feedback. For dopamine D2 receptor antagonists, it has been hypothesized that fast receptor binding kinetics cause fewer side effects, because part of the dynamics of the dopaminergic system is preserved by displacement of these antagonists. Experimental Approach Target binding kinetics of D2 receptor antagonists and signal transduction after dopamine and D2 receptor antagonist exposure were measured in vitro. These data were integrated by mechanistic modelling, taking into account competitive binding of endogenous dopamine and the antagonist, the turnover of the second messenger cAMP and negative feedback by PDE turnover. Key Results The proposed signal transduction model successfully described the cellular cAMP response for 17 D2 receptor antagonists with widely different binding kinetics. Simulation of the response to fluctuating dopamine concentrations revealed that a significant effect of the target binding kinetics on the dynamics of the signalling only occurs at endogenous dopamine concentration fluctuations with frequencies below 1 min−1. Conclusions and Implications Signal transduction and feedback are important determinants of the time course of drug effects. The effect of the D2 receptor antagonist dissociation rate constant (koff) is limited to the maximal rate of fluctuations in dopamine signalling as determined by the dopamine koff and the cAMP turnover.
Collapse
Affiliation(s)
- Wilhelmus E A de Witte
- Department of Pharmacology, Leiden Academic Centre for Drug Research, Leiden, Netherlands
| | - Joost W Versfelt
- Department of Pharmacology, Leiden Academic Centre for Drug Research, Leiden, Netherlands
| | - Maria Kuzikov
- ScreeningPort, Fraunhofer Institute for Molecular Biology and Applied Ecology, Hamburg, Germany
| | - Solene Rolland
- Global Drug Discovery, Bayer Healthcare Pharmaceuticals, Berlin, Germany
| | - Victoria Georgi
- Global Drug Discovery, Bayer Healthcare Pharmaceuticals, Berlin, Germany
| | - Philip Gribbon
- ScreeningPort, Fraunhofer Institute for Molecular Biology and Applied Ecology, Hamburg, Germany
| | - Sheraz Gul
- ScreeningPort, Fraunhofer Institute for Molecular Biology and Applied Ecology, Hamburg, Germany
| | | | - Piet Hein van der Graaf
- Department of Pharmacology, Leiden Academic Centre for Drug Research, Leiden, Netherlands.,QSP, Certara, Canterbury, UK
| | - Meindert Danhof
- Department of Pharmacology, Leiden Academic Centre for Drug Research, Leiden, Netherlands
| | - Amaury Fernández-Montalván
- Global Drug Discovery, Bayer Healthcare Pharmaceuticals, Berlin, Germany.,Servier Research Institute, Croissy-sur-Seine, France
| | - Gesa Witt
- ScreeningPort, Fraunhofer Institute for Molecular Biology and Applied Ecology, Hamburg, Germany
| | - Elizabeth C M de Lange
- Department of Pharmacology, Leiden Academic Centre for Drug Research, Leiden, Netherlands
| |
Collapse
|
21
|
Binding kinetics of cariprazine and aripiprazole at the dopamine D 3 receptor. Sci Rep 2018; 8:12509. [PMID: 30131592 PMCID: PMC6104066 DOI: 10.1038/s41598-018-30794-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/03/2018] [Indexed: 12/19/2022] Open
Abstract
The dissociation behaviours of aripiprazole and cariprazine at the human D2 and D3 receptor are evaluated. A potential correlation between kinetics and in vivo profiles, especially cariprazine’s action on negative symptoms in schizophrenia, is investigated. The binding kinetics of four ligands were indirectly evaluated. After the receptor preparations were pre-incubated with the unlabelled ligands, the dissociation was initiated with an excess of [3H]spiperone. Slow dissociation kinetics characterizes aripiprazole and cariprazine at the D2 receptor. At the D3 receptor, aripiprazole exhibits a slow monophasic dissociation, while cariprazine displays a rapid biphasic behaviour. Functional ß-arrestin assays and molecular dynamics simulations at the D3 receptor confirm a biphasic binding behaviour of cariprazine. This may influence its in vivo action, as the partial agonist could react rapidly to variations in the dopamine levels of schizophrenic patients and the ligand will not quantitatively dissociate from the receptor in one single step. With these findings novel agents may be developed that display rapid, biphasic dissociation from the D3R to further investigate this effect on in vivo profiles.
Collapse
|
22
|
Structure-kinetic relationship studies of cannabinoid CB 2 receptor agonists reveal substituent-specific lipophilic effects on residence time. Biochem Pharmacol 2018; 152:129-142. [DOI: 10.1016/j.bcp.2018.03.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/16/2018] [Indexed: 02/05/2023]
|
23
|
de Witte WEA, Rottschäfer V, Danhof M, van der Graaf PH, Peletier LA, de Lange ECM. Modelling the delay between pharmacokinetics and EEG effects of morphine in rats: binding kinetic versus effect compartment models. J Pharmacokinet Pharmacodyn 2018; 45:621-635. [PMID: 29777407 PMCID: PMC6061075 DOI: 10.1007/s10928-018-9593-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 05/02/2018] [Indexed: 01/10/2023]
Abstract
Drug–target binding kinetics (as determined by association and dissociation rate constants, kon and koff) can be an important determinant of the kinetics of drug action. However, the effect compartment model is used most frequently instead of a target binding model to describe hysteresis. Here we investigate when the drug–target binding model should be used in lieu of the effect compartment model. The utility of the effect compartment (EC), the target binding kinetics (TB) and the combined effect compartment–target binding kinetics (EC–TB) model were tested on either plasma (ECPL, TBPL and EC–TBPL) or brain extracellular fluid (ECF) (ECECF, TBECF and EC–TBECF) morphine concentrations and EEG amplitude in rats. It was also analyzed when a significant shift in the time to maximal target occupancy (TmaxTO) with increasing dose, the discriminating feature between the TB and EC model, occurs in the TB model. All TB models assumed a linear relationship between target occupancy and drug effect on the EEG amplitude. All three model types performed similarly in describing the morphine pharmacodynamics data, although the EC model provided the best statistical result. The analysis of the shift in TmaxTO (∆TmaxTO) as a result of increasing dose revealed that ∆TmaxTO is decreasing towards zero if the koff is much smaller than the elimination rate constant or if the target concentration is larger than the initial morphine concentration. The results for the morphine PKPD modelling and the analysis of ∆TmaxTO indicate that the EC and TB models do not necessarily lead to different drug effect versus time curves for different doses if a delay between drug concentrations and drug effect (hysteresis) is described. Drawing mechanistic conclusions from successfully fitting one of these two models should therefore be avoided. Since the TB model can be informed by in vitro measurements of kon and koff, a target binding model should be considered more often for mechanistic modelling purposes.
Collapse
Affiliation(s)
- Wilhelmus E A de Witte
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC, Leiden, The Netherlands
| | - Vivi Rottschäfer
- Mathematical Institute, Leiden University, 2333 CA, Leiden, The Netherlands
| | - Meindert Danhof
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC, Leiden, The Netherlands
| | - Piet H van der Graaf
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC, Leiden, The Netherlands
- Certara Quantitative Systems Pharmacology, Canterbury Innovation Centre, Canterbury, CT2 7FG, UK
| | | | - Elizabeth C M de Lange
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
24
|
De Berardis D, Rapini G, Olivieri L, Di Nicola D, Tomasetti C, Valchera A, Fornaro M, Di Fabio F, Perna G, Di Nicola M, Serafini G, Carano A, Pompili M, Vellante F, Orsolini L, Martinotti G, Di Giannantonio M. Safety of antipsychotics for the treatment of schizophrenia: a focus on the adverse effects of clozapine. Ther Adv Drug Saf 2018; 9:237-256. [PMID: 29796248 PMCID: PMC5956953 DOI: 10.1177/2042098618756261] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/10/2018] [Indexed: 12/15/2022] Open
Abstract
Clozapine, a dibenzodiazepine developed in 1961, is a multireceptorial atypical antipsychotic approved for the treatment of resistant schizophrenia. Since its introduction, it has remained the drug of choice in treatment-resistant schizophrenia, despite a wide range of adverse effects, as it is a very effective drug in everyday clinical practice. However, clozapine is not considered as a top-of-the-line treatment because it may often be difficult for some patients to tolerate as some adverse effects can be particularly bothersome (i.e. sedation, weight gain, sialorrhea etc.) and it has some other potentially dangerous and life-threatening side effects (i.e. myocarditis, seizures, agranulocytosis or granulocytopenia, gastrointestinal hypomotility etc.). As poor treatment adherence in patients with resistant schizophrenia may increase the risk of a psychotic relapse, which may further lead to impaired social and cognitive functioning, psychiatric hospitalizations and increased treatment costs, clozapine adverse effects are a common reason for discontinuing this medication. Therefore, every effort should be made to monitor and minimize these adverse effects in order to improve their early detection and management. The aim of this paper is to briefly summarize and provide an update on major clozapine adverse effects, especially focusing on those that are severe and potentially life threatening, even if most of the latter are relatively uncommon.
Collapse
Affiliation(s)
- Domenico De Berardis
- National Health Service, Department of Mental Health, Psychiatric Service of Diagnosis and Treatment, ‘G. Mazzini’ Hospital, p.zza Italia 1, 64100 Teramo, Italy
| | - Gabriella Rapini
- National Health Service, Department of Mental Health, Psychiatric Service of Diagnosis and Treatment, ‘G. Mazzini’ Hospital, Teramo, Italy
| | - Luigi Olivieri
- National Health Service, Department of Mental Health, Psychiatric Service of Diagnosis and Treatment, ‘G. Mazzini’ Hospital, Teramo, Italy
| | - Domenico Di Nicola
- National Health Service, Department of Mental Health, Psychiatric Service of Diagnosis and Treatment, ‘G. Mazzini’ Hospital, Teramo, Italy
| | - Carmine Tomasetti
- Polyedra Research Group, Teramo, Italy Department of Neuroscience, Reproductive Science and Odontostomatology, School of Medicine ‘Federico II’ Naples, Naples, Italy
| | - Alessandro Valchera
- Polyedra Research Group, Teramo, Italy Villa S. Giuseppe Hospital, Hermanas Hospitalarias, Ascoli Piceno, Italy
| | - Michele Fornaro
- Department of Neuroscience, Reproductive Science and Odontostomatology, School of Medicine ‘Federico II’ Naples, Naples, Italy
| | - Fabio Di Fabio
- Polyedra Research Group, Teramo, Italy Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy
| | - Giampaolo Perna
- Hermanas Hospitalarias, FoRiPsi, Department of Clinical Neurosciences, Villa San Benedetto Menni, Albese con Cassano, Como, Italy Department of Psychiatry and Neuropsychology, University of Maastricht, Maastricht, The Netherlands Department of Psychiatry and Behavioral Sciences, Leonard Miller School of Medicine, University of Miami, Florida, USA
| | - Marco Di Nicola
- Institute of Psychiatry and Psychology, Catholic University of Sacred Heart, Rome, Italy
| | - Gianluca Serafini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy
| | - Alessandro Carano
- National Health Service, Department of Mental Health, Psychiatric Service of Diagnosis and Treatment, Hospital ‘Madonna Del Soccorso’, San Benedetto del Tronto, Italy
| | - Maurizio Pompili
- Department of Neurosciences, Mental Health and Sensory Organs, Suicide Prevention Center, Sant’Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Federica Vellante
- Department of Neuroscience, Imaging and Clinical Science, Chair of Psychiatry, University ‘G. D’Annunzio’, Chieti, Italy
| | - Laura Orsolini
- Polyedra Research Group, Teramo, Italy Psychopharmacology, Drug Misuse and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hatfield, Herts, UK
| | - Giovanni Martinotti
- Department of Neuroscience, Imaging and Clinical Science, Chair of Psychiatry, University ‘G. D’Annunzio’, Chieti, Italy
| | - Massimo Di Giannantonio
- Department of Neuroscience, Imaging and Clinical Science, Chair of Psychiatry, University ‘G. D’Annunzio’, Chieti, Italy
| |
Collapse
|
25
|
Stoddart LA, Vernall AJ, Bouzo-Lorenzo M, Bosma R, Kooistra AJ, de Graaf C, Vischer HF, Leurs R, Briddon SJ, Kellam B, Hill SJ. Development of novel fluorescent histamine H 1-receptor antagonists to study ligand-binding kinetics in living cells. Sci Rep 2018; 8:1572. [PMID: 29371669 PMCID: PMC5785503 DOI: 10.1038/s41598-018-19714-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/03/2018] [Indexed: 01/01/2023] Open
Abstract
The histamine H1-receptor (H1R) is an important mediator of allergy and inflammation. H1R antagonists have particular clinical utility in allergic rhinitis and urticaria. Here we have developed six novel fluorescent probes for this receptor that are very effective for high resolution confocal imaging, alongside bioluminescence resonance energy transfer approaches to monitor H1R ligand binding kinetics in living cells. The latter technology exploits the opportunities provided by the recently described bright bioluminescent protein NanoLuc when it is fused to the N-terminus of a receptor. Two different pharmacophores (mepyramine or the fragment VUF13816) were used to generate fluorescent H1R antagonists conjugated via peptide linkers to the fluorophore BODIPY630/650. Kinetic properties of the probes showed wide variation, with the VUF13816 analogues having much longer H1R residence times relative to their mepyramine-based counterparts. The kinetics of these fluorescent ligands could also be monitored in membrane preparations providing new opportunities for future drug discovery applications.
Collapse
Affiliation(s)
- Leigh A Stoddart
- Division of Pharmacology Physiology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Andrea J Vernall
- School of Pharmacy, Division of Biomolecular Science and Medicinal Chemistry, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Monica Bouzo-Lorenzo
- Division of Pharmacology Physiology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Reggie Bosma
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, PO Box 7161, Amsterdam, The Netherlands
| | - Albert J Kooistra
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, PO Box 7161, Amsterdam, The Netherlands
| | - Chris de Graaf
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, PO Box 7161, Amsterdam, The Netherlands
| | - Henry F Vischer
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, PO Box 7161, Amsterdam, The Netherlands
| | - Rob Leurs
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, PO Box 7161, Amsterdam, The Netherlands
| | - Stephen J Briddon
- Division of Pharmacology Physiology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Barrie Kellam
- School of Pharmacy, Division of Biomolecular Science and Medicinal Chemistry, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, NG7 2RD, UK.
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK.
| | - Stephen J Hill
- Division of Pharmacology Physiology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK.
| |
Collapse
|
26
|
Trigo-Mouriño P, Griesinger C, Lee D. Label-free NMR-based dissociation kinetics determination. JOURNAL OF BIOMOLECULAR NMR 2017; 69:229-235. [PMID: 29143948 DOI: 10.1007/s10858-017-0150-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 11/03/2017] [Indexed: 06/07/2023]
Abstract
Understanding the dissociation of molecules is the basis to modulate interactions of biomedical interest. Optimizing drugs for dissociation rates is found to be important for their efficacy, selectivity, and safety. Here, we show an application of the high-power relaxation dispersion (RD) method to the determination of the dissociation rates of weak binding ligands from receptors. The experiment probes proton RD on the ligand and, therefore, avoids the need for any isotopic labeling. The large ligand excess eases the detection significantly. Importantly, the use of large spin-lock fields allows the detection of faster dissociation rates than other relaxation approaches. Moreover, this experimental approach allows to access directly the off-rate of the binding process without the need for analyzing a series of samples with increasing ligand saturation. The validity of the method is shown with small molecule interactions using two macromolecules, bovine serum albumin and tubulin heterodimers.
Collapse
Affiliation(s)
- Pablo Trigo-Mouriño
- Department of NMR-Based Structural Biology, Max-Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Christian Griesinger
- Department of NMR-Based Structural Biology, Max-Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Donghan Lee
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
27
|
Deganutti G, Zhukov A, Deflorian F, Federico S, Spalluto G, Cooke RM, Moro S, Mason JS, Bortolato A. Impact of protein-ligand solvation and desolvation on transition state thermodynamic properties of adenosine A 2A ligand binding kinetics. In Silico Pharmacol 2017; 5:16. [PMID: 29308352 PMCID: PMC5755719 DOI: 10.1007/s40203-017-0037-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/14/2017] [Indexed: 12/26/2022] Open
Abstract
Ligand-protein binding kinetic rates are growing in importance as parameters to consider in drug discovery and lead optimization. In this study we analysed using surface plasmon resonance (SPR) the transition state (TS) properties of a set of six adenosine A2A receptor inhibitors, belonging to both the xanthine and the triazolo-triazine scaffolds. SPR highlighted interesting differences among the ligands in the enthalpic and entropic components of the TS energy barriers for the binding and unbinding events. To better understand at a molecular level these differences, we developed suMetaD, a novel molecular dynamics (MD)-based approach combining supervised MD and metadynamics. This method allows simulation of the ligand unbinding and binding events. It also provides the system conformation corresponding to the highest energy barrier the ligand is required to overcome to reach the final state. For the six ligands evaluated in this study their TS thermodynamic properties were linked in particular to the role of water molecules in solvating/desolvating the pocket and the small molecules. suMetaD identified kinetic bottleneck conformations near the bound state position or in the vestibule area. In the first case the barrier is mainly enthalpic, requiring the breaking of strong interactions with the protein. In the vestibule TS location the kinetic bottleneck is instead mainly of entropic nature, linked to the solvent behaviour.
Collapse
Affiliation(s)
- Giuseppe Deganutti
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, Padua, Italy
| | - Andrei Zhukov
- Heptares Therapeutics Ltd., BioPark, Broadwater Road, Welwyn Garden City, Herts AL7 3AX UK
| | - Francesca Deflorian
- Heptares Therapeutics Ltd., BioPark, Broadwater Road, Welwyn Garden City, Herts AL7 3AX UK
| | - Stephanie Federico
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Piazzale Europa, 34127 Trieste, Italy
| | - Giampiero Spalluto
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Piazzale Europa, 34127 Trieste, Italy
| | - Robert M. Cooke
- Heptares Therapeutics Ltd., BioPark, Broadwater Road, Welwyn Garden City, Herts AL7 3AX UK
| | - Stefano Moro
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, Padua, Italy
| | - Jonathan S. Mason
- Heptares Therapeutics Ltd., BioPark, Broadwater Road, Welwyn Garden City, Herts AL7 3AX UK
| | - Andrea Bortolato
- Heptares Therapeutics Ltd., BioPark, Broadwater Road, Welwyn Garden City, Herts AL7 3AX UK
| |
Collapse
|
28
|
Bosma R, Witt G, Vaas LAI, Josimovic I, Gribbon P, Vischer HF, Gul S, Leurs R. The Target Residence Time of Antihistamines Determines Their Antagonism of the G Protein-Coupled Histamine H1 Receptor. Front Pharmacol 2017; 8:667. [PMID: 29033838 PMCID: PMC5627017 DOI: 10.3389/fphar.2017.00667] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/07/2017] [Indexed: 11/13/2022] Open
Abstract
The pharmacodynamics of drug-candidates is often optimized by metrics that describe target binding (Kd or Ki value) or target modulation (IC50). However, these metrics are determined at equilibrium conditions, and consequently information regarding the onset and offset of target engagement and modulation is lost. Drug-target residence time is a measure for the lifetime of the drug-target complex, which has recently been receiving considerable interest, as target residence time is shown to have prognostic value for the in vivo efficacy of several drugs. In this study, we have investigated the relation between the increased residence time of antihistamines at the histamine H1 receptor (H1R) and the duration of effective target-inhibition by these antagonists. Hela cells, endogenously expressing low levels of the H1R, were incubated with a series of antihistamines and dissociation was initiated by washing away the unbound antihistamines. Using a calcium-sensitive fluorescent dye and a label free, dynamic mass redistribution based assay, functional recovery of the H1R responsiveness was measured by stimulating the cells with histamine over time, and the recovery was quantified as the receptor recovery time. Using these assays, we determined that the receptor recovery time for a set of antihistamines differed more than 40-fold and was highly correlated to their H1R residence times, as determined with competitive radioligand binding experiments to the H1R in a cell homogenate. Thus, the receptor recovery time is proposed as a cell-based and physiologically relevant metric for the lead optimization of G protein-coupled receptor antagonists, like the H1R antagonists. Both, label-free or real-time, classical signaling assays allow an efficient and physiologically relevant determination of kinetic properties of drug molecules.
Collapse
Affiliation(s)
- Reggie Bosma
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| | - Gesa Witt
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Lea A I Vaas
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Ivana Josimovic
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| | - Philip Gribbon
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Henry F Vischer
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Rob Leurs
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| |
Collapse
|
29
|
Kharkwal G, Brami-Cherrier K, Lizardi-Ortiz JE, Nelson AB, Ramos M, Del Barrio D, Sulzer D, Kreitzer AC, Borrelli E. Parkinsonism Driven by Antipsychotics Originates from Dopaminergic Control of Striatal Cholinergic Interneurons. Neuron 2017; 91:67-78. [PMID: 27387649 DOI: 10.1016/j.neuron.2016.06.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 04/11/2016] [Accepted: 05/10/2016] [Indexed: 02/03/2023]
Abstract
Typical antipsychotics can cause disabling side effects. Specifically, antagonism of D2R signaling by the typical antipsychotic haloperidol induces parkinsonism in humans and catalepsy in rodents. Striatal dopamine D2 receptors (D2R) are major regulators of motor activity through their signaling on striatal projection neurons and interneurons. We show that D2R signaling on cholinergic interneurons contributes to an in vitro pause in firing of these otherwise tonically active neurons and to the striatal dopamine/acetylcholine balance. The selective ablation of D2R from cholinergic neurons allows discrimination between the motor-reducing and cataleptic effects of antipsychotics. The cataleptic effect of antipsychotics is triggered by blockade of D2R on cholinergic interneurons and the consequent increase of acetylcholine signaling on striatal projection neurons. These studies illuminate the critical role of D2R-mediated signaling in regulating the activity of striatal cholinergic interneurons and the mechanisms of typical antipsychotic side effects.
Collapse
Affiliation(s)
- Geetika Kharkwal
- Department of Microbiology & Molecular Genetics, U904 INSERM, University of California, Irvine, Irvine, CA 92697, USA
| | - Karen Brami-Cherrier
- Department of Microbiology & Molecular Genetics, U904 INSERM, University of California, Irvine, Irvine, CA 92697, USA
| | - José E Lizardi-Ortiz
- Departments of Neurology and Pharmacology, Columbia University, New York, NY 10032, USA
| | - Alexandra B Nelson
- The Gladstone Institutes, San Francisco, CA 94158, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Maria Ramos
- Department of Microbiology & Molecular Genetics, U904 INSERM, University of California, Irvine, Irvine, CA 92697, USA
| | - Daniel Del Barrio
- Department of Microbiology & Molecular Genetics, U904 INSERM, University of California, Irvine, Irvine, CA 92697, USA
| | - David Sulzer
- Departments of Neurology and Pharmacology, Columbia University, New York, NY 10032, USA
| | | | - Emiliana Borrelli
- Department of Microbiology & Molecular Genetics, U904 INSERM, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
30
|
Rath JJG, Deen MEJ, van Houten H, de Bruijn SFTM, van Gerven J, Mul D. Antipsychotic-induced hyperprolactinemia in Tourette syndrome. Ther Adv Psychopharmacol 2017; 7:201-205. [PMID: 28740637 PMCID: PMC5505441 DOI: 10.1177/2045125317705012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 01/25/2017] [Indexed: 01/04/2023] Open
Abstract
For many years, Tourette syndrome (TS) was considered to be a rare disorder, but tics and TS are now recognized as fairly common childhood-onset conditions. Children and adolescents with TS are frequently treated with antipsychotics, either as monotherapy or in combination with psychostimulants, melatonin and selective serotonin reuptake inhibitors (SSRIs). Antipsychotics are most often used in schizophrenia and related psychotic disorders, and in these conditions hyperprolactinemia is one of the most common adverse effects associated with antipsychotics, occurring in 40-50% of patients. We describe two patients with TS who experienced antipsychotic-induced hyperprolactinemia. Treatment options generally consist of dose reduction or switching from typical to atypical antipsychotics. However, diminishing dosages can lead to exacerbations of tics. Also, not all atypical antipsychotics have the same pharmacologic properties required to normalize prolactin levels. The choice of treatment may also be affected by the patient's age and sex. These factors are discussed in relation to these cases, and illustrated by the results of therapeutic interventions over the years.
Collapse
Affiliation(s)
- Judith J G Rath
- Department of Neurology, LangeLand Hospital, Toneellaan 1, 2725 NA Zoetermeer, The Netherlands
| | - Marlies E J Deen
- Department of Pediatrics, Haga Teaching Hospital, The Hague, The Netherlands
| | - Hessel van Houten
- Department of Internal Medicine, Haga Teaching Hospital, The Hague, The Netherlands
| | | | | | - Dick Mul
- Diabeter, Center for Pediatric and Adolescent Diabetes Care and Research, Rotterdam, The Netherlands
| |
Collapse
|
31
|
Vanderheyden PML, Benachour N. Influence of the cellular environment on ligand binding kinetics at membrane-bound targets. Bioorg Med Chem Lett 2017; 27:3621-3628. [PMID: 28666735 DOI: 10.1016/j.bmcl.2017.06.051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/13/2017] [Accepted: 06/19/2017] [Indexed: 10/19/2022]
Abstract
While historically 'in vitro' binding data were obtained by analyzing equilibrium experiments, kinetic data are increasingly appreciated to provide information on the time a particular compound remains bound to its target. This information is of biological importance to understand the molecular mechanism of a drug not only to evaluate the time a particular receptor/enzyme is blocked in the case of antagonists/inhibitors but also to investigate its contribution to the efficacy to mediate signaling in the case of agonists. There is accumulating evidence that many drugs binding to either membrane-bound receptors or enzymes are found to display long duration of action which can be ascribed to slow dissociation from their target proteins. In the present review three such examples are discussed which encompass ligands that bind to membrane-bound proteins and from which it appears that the tight binding kinetics is influenced by the cellular/membrane environment of the target protein.
Collapse
Affiliation(s)
- Patrick M L Vanderheyden
- Research Group of Molecular and Biochemical Pharmacology, Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, VUB-MBFA, Pleinlaan 2, B-1050 Brussels, Belgium.
| | - Nerdjes Benachour
- Research Group of Molecular and Biochemical Pharmacology, Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, VUB-MBFA, Pleinlaan 2, B-1050 Brussels, Belgium
| |
Collapse
|
32
|
Schuetz DA, de Witte WEA, Wong YC, Knasmueller B, Richter L, Kokh DB, Sadiq SK, Bosma R, Nederpelt I, Heitman LH, Segala E, Amaral M, Guo D, Andres D, Georgi V, Stoddart LA, Hill S, Cooke RM, De Graaf C, Leurs R, Frech M, Wade RC, de Lange ECM, IJzerman AP, Müller-Fahrnow A, Ecker GF. Kinetics for Drug Discovery: an industry-driven effort to target drug residence time. Drug Discov Today 2017; 22:896-911. [PMID: 28412474 DOI: 10.1016/j.drudis.2017.02.002] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 01/24/2017] [Accepted: 02/17/2017] [Indexed: 01/05/2023]
Abstract
A considerable number of approved drugs show non-equilibrium binding characteristics, emphasizing the potential role of drug residence times for in vivo efficacy. Therefore, a detailed understanding of the kinetics of association and dissociation of a target-ligand complex might provide crucial insight into the molecular mechanism-of-action of a compound. This deeper understanding will help to improve decision making in drug discovery, thus leading to a better selection of interesting compounds to be profiled further. In this review, we highlight the contributions of the Kinetics for Drug Discovery (K4DD) Consortium, which targets major open questions related to binding kinetics in an industry-driven public-private partnership.
Collapse
Affiliation(s)
- Doris A Schuetz
- Department of Pharmaceutical Chemistry, University of Vienna, UZA 2, Althanstrasse 14, 1090 Vienna, Austria
| | | | - Yin Cheong Wong
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Bernhard Knasmueller
- Department of Pharmaceutical Chemistry, University of Vienna, UZA 2, Althanstrasse 14, 1090 Vienna, Austria
| | - Lars Richter
- Department of Pharmaceutical Chemistry, University of Vienna, UZA 2, Althanstrasse 14, 1090 Vienna, Austria
| | - Daria B Kokh
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), Schloß-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany
| | - S Kashif Sadiq
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), Schloß-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany
| | - Reggie Bosma
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, P.O. Box 7161, 1007 MC Amsterdam, The Netherlands
| | - Indira Nederpelt
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, Leiden, Einsteinweg 55, Leiden, 2300RA, The Netherlands
| | - Laura H Heitman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, Leiden, Einsteinweg 55, Leiden, 2300RA, The Netherlands
| | - Elena Segala
- Heptares Therapeutics,Biopark, Broadwater Road, Welwyn Garden City, Hertfordshire, AL7 3AX, UK
| | - Marta Amaral
- Discovery Technologies, Merck KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany; Instituto de Biologia Experimental e Tecnológica, Avenida da República, Estação Agronómica Nacional, 2780-157 Oeiras, Portugal
| | - Dong Guo
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, Leiden, Einsteinweg 55, Leiden, 2300RA, The Netherlands
| | - Dorothee Andres
- Bayer AG, Drug Discovery, Pharmaceuticals, Lead Discovery Berlin, Müllerstr. 178, 13353 Berlin, Germany
| | - Victoria Georgi
- Bayer AG, Drug Discovery, Pharmaceuticals, Lead Discovery Berlin, Müllerstr. 178, 13353 Berlin, Germany
| | - Leigh A Stoddart
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Steve Hill
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Robert M Cooke
- Heptares Therapeutics,Biopark, Broadwater Road, Welwyn Garden City, Hertfordshire, AL7 3AX, UK
| | - Chris De Graaf
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, P.O. Box 7161, 1007 MC Amsterdam, The Netherlands
| | - Rob Leurs
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, P.O. Box 7161, 1007 MC Amsterdam, The Netherlands
| | - Matthias Frech
- Discovery Technologies, Merck KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Rebecca C Wade
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), Schloß-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany; Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Alliance, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Im Neuenheimer Feld 205, 69120 Heidelberg, Germany
| | - Elizabeth Cunera Maria de Lange
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Adriaan P IJzerman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, Leiden, Einsteinweg 55, Leiden, 2300RA, The Netherlands
| | - Anke Müller-Fahrnow
- Bayer AG, Drug Discovery, Pharmaceuticals, Lead Discovery Berlin, Müllerstr. 178, 13353 Berlin, Germany
| | - Gerhard F Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, UZA 2, Althanstrasse 14, 1090 Vienna, Austria.
| |
Collapse
|
33
|
Estimation of kinetic and thermodynamic ligand-binding parameters using computational strategies. Future Med Chem 2017; 9:507-523. [DOI: 10.4155/fmc-2016-0224] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Kinetic and thermodynamic ligand–protein binding parameters are gaining growing importance as key information to consider in drug discovery. The determination of the molecular structures, using particularly x-ray and NMR techniques, is crucial for understanding how a ligand recognizes its target in the final binding complex. However, for a better understanding of the recognition processes, experimental studies of ligand–protein interactions are needed. Even though several techniques can be used to investigate both thermodynamic and kinetic profiles for a ligand–protein complex, these procedures are very often laborious, time consuming and expensive. In the last 10 years, computational approaches have enormous potential in providing insights into each of the above effects and in parsing their contributions to the changes in both kinetic and thermodynamic binding parameters. The main purpose of this review is to summarize the state of the art of computational strategies for estimating the kinetic and thermodynamic parameters of a ligand–protein binding.
Collapse
|
34
|
Bernetti M, Cavalli A, Mollica L. Protein-ligand (un)binding kinetics as a new paradigm for drug discovery at the crossroad between experiments and modelling. MEDCHEMCOMM 2017; 8:534-550. [PMID: 30108770 PMCID: PMC6072069 DOI: 10.1039/c6md00581k] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/25/2017] [Indexed: 12/14/2022]
Abstract
In the last three decades, protein and nucleic acid structure determination and comprehension of the mechanisms, leading to their physiological and pathological functions, have become a cornerstone of biomedical sciences. A deep understanding of the principles governing the fates of cells and tissue at the molecular level has been gained over the years, offering a solid basis for the rational design of drugs aimed at the pharmacological treatment of numerous diseases. Historically, affinity indicators (i.e. Kd and IC50/EC50) have been assumed to be valid indicators of the in vivo efficacy of a drug. However, recent studies pointed out that the kinetics of the drug-receptor binding process could be as important or even more important than affinity in determining the drug efficacy. This eventually led to a growing interest in the characterisation and prediction of the rate constants of protein-ligand association and dissociation. For instance, a drug with a longer residence time can kinetically select a given receptor over another, even if the affinity for both receptors is comparable, thus increasing its therapeutic index. Therefore, understanding the molecular features underlying binding and unbinding processes is of central interest towards the rational control of drug binding kinetics. In this review, we report the theoretical framework behind protein-ligand association and highlight the latest advances in the experimental and computational approaches exploited to investigate the binding kinetics.
Collapse
Affiliation(s)
- M Bernetti
- Department of Pharmacy and Biotechnology , University of Bologna , via Belmeloro 6 , 40126 Bologna , Italy
- CompuNet , Istituto Italiano di Tecnologia , via Morego 30 , 16163 Genova , Italy .
| | - A Cavalli
- Department of Pharmacy and Biotechnology , University of Bologna , via Belmeloro 6 , 40126 Bologna , Italy
- CompuNet , Istituto Italiano di Tecnologia , via Morego 30 , 16163 Genova , Italy .
| | - L Mollica
- CompuNet , Istituto Italiano di Tecnologia , via Morego 30 , 16163 Genova , Italy .
| |
Collapse
|
35
|
Piwowarska J, Radziwoń-Zaleska M, Dmochowska M, Szepietowska E, Matsumoto H, Sygitowicz G, Pilc A, Łukaszkiewicz J. The usefulness of monitored therapy using Clozapine concentration in the blood serum for determining drug dose in Polish schizophrenic patients. Pharmacol Rep 2016; 68:1120-1125. [DOI: 10.1016/j.pharep.2016.06.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 06/23/2016] [Accepted: 06/24/2016] [Indexed: 11/27/2022]
|
36
|
Zanatta G, Della Flora Nunes G, Bezerra EM, da Costa RF, Martins A, Caetano EWS, Freire VN, Gottfried C. Two Binding Geometries for Risperidone in Dopamine D3 Receptors: Insights on the Fast-Off Mechanism through Docking, Quantum Biochemistry, and Molecular Dynamics Simulations. ACS Chem Neurosci 2016; 7:1331-1347. [PMID: 27434874 DOI: 10.1021/acschemneuro.6b00074] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Risperidone is an atypical antipsychotic used in the treatment of schizophrenia and of symptoms of irritability associated with autism spectrum disorder (ASD). Its main action mechanism is the blockade of D2-like receptors acting over positive and negative symptoms of schizophrenia with small risk of extrapyramidal symptoms (EPS) at doses corresponding to low/moderate D2 occupancy. Such a decrease in the side effect incidence can be associated with its fast unbinding from D2 receptors in the nigrostriatal region allowing the recovery of dopamine signaling pathways. We performed docking essays using risperidone and the D3 receptor crystallographic data and results suggested two possible distinct orientations for risperidone at the binding pocket. Orientation 1 is more close to the opening of the binding site and has the 6-fluoro-1,2 benzoxazole fragment toward the bottom of the D3 receptor cleft, while orientation 2 is deeper inside the binding pocket with the same fragment toward to the receptor surface. In order to unveil the implications of these two binding orientations, classical molecular dynamics and quantum biochemistry computations within the density functional theory formalism and the molecular fractionation with conjugate caps framework were performed. Quantum mechanics/molecular mechanics suggests that orientation 2 (considering the contribution of Glu90) is slightly more energetically stable than orientation 1 with the main contribution coming from residue Asp110. The residue Glu90, positioned at the opening of the binding site, is closer to orientation 1 than 2, suggesting that it may have a key role in stability through attractive interaction with risperidone. Therefore, although orientations 1 and 2 are both likely to occur, we suggest that the occurrence of the first may contribute to the reduction of side effects in patients taking risperidone due to the reduction of dopamine receptor occupancy in the nigrostriatal region through a mechanism of fast dissociation. The atypical effect may be obtained simply by either delaying D3R full blockage by spatial hindrance of orientation 1 at the binding site or through an effective blockade followed by orientation 1 fast dissociation. While the molecular interpretation suggested in this work shed some light on the potential molecular mechanisms accounting for the reduced extrapyramidal symptoms observed during risperidone treatment, further studies are necessary in order to evaluate the implications of both orientations during the receptor activation/inhibition. Altogether these data highlight important hot spots in the dopamine receptor binding site bringing relevant information for the development of novel/derivative agents with atypical profile.
Collapse
Affiliation(s)
- Geancarlo Zanatta
- Department of Biochemistry, Federal University of Rio Grande do Sul, 90035-003 Porto
Alegre, RS Brazil
| | - Gustavo Della Flora Nunes
- Department of Biochemistry, Federal University of Rio Grande do Sul, 90035-003 Porto
Alegre, RS Brazil
| | - Eveline M. Bezerra
- Post-graduate Program in Pharmaceutical Sciences, Pharmacy Faculty, Federal University of Ceará, 60430-372 Fortaleza, CE Brazil
| | - Roner F. da Costa
- Department of Physics, Universidade Federal Rural do Semi-Árido, 59780-000 Caraúbas, RN Brazil
| | - Alice Martins
- Post-graduate Program in Pharmaceutical Sciences, Pharmacy Faculty, Federal University of Ceará, 60430-372 Fortaleza, CE Brazil
| | - Ewerton W. S. Caetano
- Federal Institute of Education, Science and Technology, 60040-531 Fortaleza, CE Brazil
| | - Valder N. Freire
- Department of Physics, Federal University of Ceará, 60455-760 Fortaleza, CE Brazil
| | - Carmem Gottfried
- Department of Biochemistry, Federal University of Rio Grande do Sul, 90035-003 Porto
Alegre, RS Brazil
| |
Collapse
|
37
|
Vauquelin G. Cell membranes… and how long drugs may exert beneficial pharmacological activity in vivo. Br J Clin Pharmacol 2016; 82:673-82. [PMID: 27135195 PMCID: PMC5338106 DOI: 10.1111/bcp.12996] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/28/2016] [Accepted: 04/28/2016] [Indexed: 12/14/2022] Open
Abstract
The time course of the beneficial pharmacological effect of a drug has long been considered to depend merely on the temporal fluctuation of its free concentration. Only in the last decade has it become widely accepted that target-binding kinetics can also affect in vivo pharmacological activity. Although current reviews still essentially focus on genuine dissociation rates, evidence is accumulating that additional micro-pharmacokinetic (PK) and -pharmacodynamic (PD) mechanisms, in which the cell membrane plays a central role, may also increase the residence time of a drug on its target. The present review provides a compilation of otherwise widely dispersed information on this topic. The cell membrane can intervene in drug binding via the following three major mechanisms: (i) by acting as a sink/repository for the drug; (ii) by modulating the conformation of the drug and even by participating in the binding process; and (iii) by facilitating the approach (and rebinding) of the drug to the target. To highlight these mechanisms, we focus on drugs that are currently used in clinical therapy, such as the antihypertensive angiotensin II type 1 receptor antagonist candesartan, the atypical antipsychotic agent clozapine and the bronchodilator salmeterol. Although the role of cell membranes in PK-PD modelling is gaining increasing interest, many issues remain unresolved. It is likely that novel biophysical and computational approaches will provide improved insights in the near future.
Collapse
Affiliation(s)
- Georges Vauquelin
- Department Molecular and Biochemical PharmacologyVrije Universiteit BrusselBrusselsBelgium
| |
Collapse
|
38
|
Guo D, Heitman LH, IJzerman AP. Kinetic Aspects of the Interaction between Ligand and G Protein-Coupled Receptor: The Case of the Adenosine Receptors. Chem Rev 2016; 117:38-66. [DOI: 10.1021/acs.chemrev.6b00025] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Dong Guo
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Laura H. Heitman
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Adriaan P. IJzerman
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| |
Collapse
|
39
|
Thomas T, Fang Y, Yuriev E, Chalmers DK. Ligand Binding Pathways of Clozapine and Haloperidol in the Dopamine D2 and D3 Receptors. J Chem Inf Model 2016; 56:308-21. [PMID: 26690887 DOI: 10.1021/acs.jcim.5b00457] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The binding of a small molecule ligand to its protein target is most often characterized by binding affinity and is typically viewed as an on/off switch. The more complex reality is that binding involves the ligand passing through a series of intermediate states between the solution phase and the fully bound pose. We have performed a set of 29 unbiased molecular dynamics simulations to model the binding pathways of the dopamine receptor antagonists clozapine and haloperidol binding to the D2 and D3 dopamine receptors. Through these simulations we have captured the binding pathways of clozapine and haloperidol from the extracellular vestibule to the orthosteric binding site and thereby, we also predict the bound pose of each ligand. These are the first long time scale simulations of haloperidol or clozapine binding to dopamine receptors. From these simulations, we have identified several important stages in the binding pathway, including the involvement of Tyr7.35 in a "handover" mechanism that transfers the ligand between the extracellular vestibule and Asp3.32. We have also performed interaction and cluster analyses to determine differences in binding pathways between the D2 and D3 receptors and identified metastable states that may be of use in drug design.
Collapse
Affiliation(s)
- Trayder Thomas
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University , 381 Royal Pde, Parkville, Victoria 3052, Australia
| | - Yu Fang
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University , 381 Royal Pde, Parkville, Victoria 3052, Australia
| | - Elizabeth Yuriev
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University , 381 Royal Pde, Parkville, Victoria 3052, Australia
| | - David K Chalmers
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University , 381 Royal Pde, Parkville, Victoria 3052, Australia
| |
Collapse
|
40
|
Vitucci D, Di Giorgio A, Napolitano F, Pelosi B, Blasi G, Errico F, Attrotto MT, Gelao B, Fazio L, Taurisano P, Di Maio A, Marsili V, Pasqualetti M, Bertolino A, Usiello A. Rasd2 Modulates Prefronto-Striatal Phenotypes in Humans and 'Schizophrenia-Like Behaviors' in Mice. Neuropsychopharmacology 2016; 41:916-27. [PMID: 26228524 PMCID: PMC4707838 DOI: 10.1038/npp.2015.228] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/03/2015] [Accepted: 07/25/2015] [Indexed: 12/18/2022]
Abstract
Rasd2 is a thyroid hormone target gene, which encodes for a GTP-binding protein enriched in the striatum where, among other functions, it modulates dopaminergic neurotransmission. Here we report that human RASD2 mRNA is abundant in putamen, but it also occurs in the cerebral cortex, with a distinctive expression pattern that differs from that present in rodents. Consistent with its localization, we found that a genetic variation in RASD2 (rs6518956) affects postmortem prefrontal mRNA expression in healthy humans and is associated with phenotypes of relevance to schizophrenia, including prefrontal and striatal grey matter volume and physiology during working memory, as measured with magnetic resonance imaging. Interestingly, quantitative real-time PCR analysis indicated that RASD2 mRNA is slightly reduced in postmortem prefrontal cortex of patients with schizophrenia. In the attempt to uncover the neurobiological substrates associated with Rasd2 activity, we used knockout mice to analyze the in vivo influence of this G-protein on the prepulse inhibition of the startle response and psychotomimetic drug-related behavioral response. Data showed that Rasd2 mutants display deficits in basal prepulse inhibition that, in turn, exacerbate gating disruption under psychotomimetic drug challenge. Furthermore, we documented that lack of Rasd2 strikingly enhances the behavioral sensitivity to motor stimulation elicited by amphetamine and phencyclidine. Based on animal model data, along with the finding that RASD2 influences prefronto-striatal phenotypes in healthy humans, we suggest that genetic mutation or reduced levels of this G-protein might have a role in cerebral circuitry dysfunction underpinning exaggerated psychotomimetic drugs responses and development of specific biological phenotypes linked to schizophrenia.
Collapse
Affiliation(s)
- Daniela Vitucci
- Ceinge Biotecnologie Avanzate, Naples, Italy,Dipartimento di Scienze Motorie e del Benessere DiSMeB, Università degli Studi di Napoli Parthenope, Naples, Italy
| | - Annabella Di Giorgio
- Istituto di Ricovero e Cura a Carattere Scientifico ‘Casa Sollievo della Sofferenza', Foggia, Italy
| | - Francesco Napolitano
- Ceinge Biotecnologie Avanzate, Naples, Italy,Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II', Naples, Italy
| | - Barbara Pelosi
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - Giuseppe Blasi
- Group of Psychiatric Neuroscience, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro', Bari, Italy
| | - Francesco Errico
- Ceinge Biotecnologie Avanzate, Naples, Italy,Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II', Naples, Italy
| | - Maria Teresa Attrotto
- Group of Psychiatric Neuroscience, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro', Bari, Italy
| | - Barbara Gelao
- Group of Psychiatric Neuroscience, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro', Bari, Italy
| | - Leonardo Fazio
- Group of Psychiatric Neuroscience, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro', Bari, Italy
| | - Paolo Taurisano
- Group of Psychiatric Neuroscience, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro', Bari, Italy
| | | | | | - Massimo Pasqualetti
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy,Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto (Trento), Italy
| | - Alessandro Bertolino
- Group of Psychiatric Neuroscience, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro', Bari, Italy,pRED, Neuroscience DTA, Hoffmann-La Roche, Basel, Switzerland,Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro', Piazza G. Cesare 11, Bari 70124, Italy, Tel: +39 0805478572, Fax: +39 0805593172,
| | - Alessandro Usiello
- Ceinge Biotecnologie Avanzate, Naples, Italy,Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples (SUN), Caserta, Italy,Ceinge Biotecnologie Avanzate, Via G. Salvatore 486, Naples 80145, Italy, Tel: +39 0813737899, Fax: +39 0813737808. E-mail:
| |
Collapse
|
41
|
Abstract
The drug-target residence time model was first introduced in 2006 and has been broadly adopted across the chemical biology, biotechnology and pharmaceutical communities. While traditional in vitro methods view drug-target interactions exclusively in terms of equilibrium affinity, the residence time model takes into account the conformational dynamics of target macromolecules that affect drug binding and dissociation. The key tenet of this model is that the lifetime (or residence time) of the binary drug-target complex, and not the binding affinity per se, dictates much of the in vivo pharmacological activity. Here, this model is revisited and key applications of it over the past 10 years are highlighted.
Collapse
|
42
|
de Witte WEA, Wong YC, Nederpelt I, Heitman LH, Danhof M, van der Graaf PH, Gilissen RAHJ, de Lange ECM. Mechanistic models enable the rational use of in vitro drug-target binding kinetics for better drug effects in patients. Expert Opin Drug Discov 2015; 11:45-63. [PMID: 26484747 DOI: 10.1517/17460441.2016.1100163] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Drug-target binding kinetics are major determinants of the time course of drug action for several drugs, as clearly described for the irreversible binders omeprazole and aspirin. This supports the increasing interest to incorporate newly developed high-throughput assays for drug-target binding kinetics in drug discovery. A meaningful application of in vitro drug-target binding kinetics in drug discovery requires insight into the relation between in vivo drug effect and in vitro measured drug-target binding kinetics. AREAS COVERED In this review, the authors discuss both the relation between in vitro and in vivo measured binding kinetics and the relation between in vivo binding kinetics, target occupancy and effect profiles. EXPERT OPINION More scientific evidence is required for the rational selection and development of drug-candidates on the basis of in vitro estimates of drug-target binding kinetics. To elucidate the value of in vitro binding kinetics measurements, it is necessary to obtain information on system-specific properties which influence the kinetics of target occupancy and drug effect. Mathematical integration of this information enables the identification of drug-specific properties which lead to optimal target occupancy and drug effect in patients.
Collapse
Affiliation(s)
- Wilhelmus E A de Witte
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Yin Cheong Wong
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Indira Nederpelt
- b Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Laura H Heitman
- b Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Meindert Danhof
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Piet H van der Graaf
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Ron A H J Gilissen
- c A Division of Janssen Pharmaceutica N.V., Janssen Research and Development , Turnhoutseweg 30, Beerse 2340 , Belgium
| | - Elizabeth C M de Lange
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| |
Collapse
|
43
|
Vauquelin G, Van Liefde I, Swinney DC. Radioligand binding to intact cells as a tool for extended drug screening in a representative physiological context. DRUG DISCOVERY TODAY. TECHNOLOGIES 2015; 17:28-34. [PMID: 26724334 DOI: 10.1016/j.ddtec.2015.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 09/04/2015] [Accepted: 09/09/2015] [Indexed: 06/05/2023]
Abstract
Radioligand binding assays on intact cells offer distinct advantages to those on membrane suspensions. Major pharmacological properties like drug affinity and binding kinetics are more physiologically relevant. Complex mechanisms can be studied with a wider choice of experimental approaches and so provide insights into induced-fit type binding, receptor internalisation and even into pharmacomicrokinetic phenomena like drug rebinding and partitioning into the membrane. Hence, intact cell binding constitutes a valuable addition to the pharmacologist's toolbox.
Collapse
Affiliation(s)
- Georges Vauquelin
- Dept. of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium.
| | - Isabelle Van Liefde
- Dept. of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - David C Swinney
- Institute for Rare and Neglected Diseases Drug Discovery, 897 Independence Ave, Suite 2C, Mountain View, CA 94043, United States
| |
Collapse
|
44
|
Pompeu TET, Monteiro do Monte F, Bosier B, Fraga CAM, Barreiro EJ, Menegatti R, Hermans E, Noël F. Partial agonism and fast dissociation of LASSBio-579 at dopamine D2 receptor. Prog Neuropsychopharmacol Biol Psychiatry 2015; 62:1-6. [PMID: 25891249 DOI: 10.1016/j.pnpbp.2015.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 03/30/2015] [Accepted: 04/11/2015] [Indexed: 01/09/2023]
Abstract
In an attempt to better understand the molecular mechanism of action of the antipsychotic lead LASSBio-579 and of its main metabolite LQFM 037, the aim of this work was to evaluate their intrinsic activity and binding kinetics at the dopamine D2 receptor. In transfected HEK cells expressing the D2L receptor under an inducible promoter, LASSBio-579 and LQFM 037, but not clozapine, behaved as weak partial agonists in [(35)S]-GTPγS binding assays performed in optimized conditions previously shown to evidence the partial agonist profile of aripiprazole. Besides, data obtained in radioligand competition assays on rat striatal membranes suggested a rapid association to and dissociation from the D2-like receptors. Using the kinetic rate index based on the strategy of the dual-point competition association assay, we showed that our compounds share a similar kinetic profile with clozapine, distinct from the typical antipsychotic haloperidol. These two characteristics could contribute to the atypical-like profile observed after administration of LASSBio-579 to rodents, in models of positive and negative symptoms of schizophrenia.
Collapse
Affiliation(s)
- Thais Emanoelle T Pompeu
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Sala J1-17, CEP 21941-912 Ilha do Fundão, Rio de Janeiro, Brazil; Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Sala K2-27, CEP 21941-912 Ilha do Fundão, Rio de Janeiro, Brazil
| | - Fernando Monteiro do Monte
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Sala J1-17, CEP 21941-912 Ilha do Fundão, Rio de Janeiro, Brazil; Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Sala K2-27, CEP 21941-912 Ilha do Fundão, Rio de Janeiro, Brazil
| | - Barbara Bosier
- Institute of Neuroscience (IoNS), Group of Neuropharmacology, Université Catholique de Louvain, Brussels, Belgium
| | - Carlos A M Fraga
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Sala K2-27, CEP 21941-912 Ilha do Fundão, Rio de Janeiro, Brazil; Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco Bss 022, CEP 21944-971 Ilha do Fundão, Rio de Janeiro, Brazil
| | - Eliezer J Barreiro
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Sala K2-27, CEP 21941-912 Ilha do Fundão, Rio de Janeiro, Brazil; Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco Bss 022, CEP 21944-971 Ilha do Fundão, Rio de Janeiro, Brazil
| | - Ricardo Menegatti
- Laboratório de Bioconversão, LabMol and Laboratório de Química Farmacêutica, Medicinal, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Emmanuel Hermans
- Institute of Neuroscience (IoNS), Group of Neuropharmacology, Université Catholique de Louvain, Brussels, Belgium
| | - François Noël
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Sala J1-17, CEP 21941-912 Ilha do Fundão, Rio de Janeiro, Brazil; Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Sala K2-27, CEP 21941-912 Ilha do Fundão, Rio de Janeiro, Brazil.
| |
Collapse
|
45
|
Guo D, Heitman LH, IJzerman AP. The Role of Target Binding Kinetics in Drug Discovery. ChemMedChem 2015; 10:1793-6. [DOI: 10.1002/cmdc.201500310] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Indexed: 12/31/2022]
Affiliation(s)
- Dong Guo
- Division of Medicinal Chemistry; Leiden Academic Centre for Drug Research (LACDR); P.O. Box 9502 2300 RA Leiden the Netherlands
| | - Laura H. Heitman
- Division of Medicinal Chemistry; Leiden Academic Centre for Drug Research (LACDR); P.O. Box 9502 2300 RA Leiden the Netherlands
| | - Adriaan P. IJzerman
- Division of Medicinal Chemistry; Leiden Academic Centre for Drug Research (LACDR); P.O. Box 9502 2300 RA Leiden the Netherlands
| |
Collapse
|
46
|
Affiliation(s)
- Nahla Mahgoub
- Weil Cornell University, Westchester Division, White Plains, NY
| |
Collapse
|
47
|
Bradshaw JM, McFarland JM, Paavilainen VO, Bisconte A, Tam D, Phan VT, Romanov S, Finkle D, Shu J, Patel V, Ton T, Li X, Loughhead DG, Nunn PA, Karr DE, Gerritsen ME, Funk JO, Owens TD, Verner E, Brameld KA, Hill RJ, Goldstein DM, Taunton J. Prolonged and tunable residence time using reversible covalent kinase inhibitors. Nat Chem Biol 2015; 11:525-31. [PMID: 26006010 PMCID: PMC4472506 DOI: 10.1038/nchembio.1817] [Citation(s) in RCA: 283] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 04/13/2015] [Indexed: 12/11/2022]
Abstract
Drugs with prolonged, on-target residence time often show superior efficacy, yet general strategies for optimizing drug-target residence time are lacking. Here, we demonstrate progress toward this elusive goal by targeting a noncatalytic cysteine in Bruton's tyrosine kinase (BTK) with reversible covalent inhibitors. Utilizing an inverted orientation of the cysteine-reactive cyanoacrylamide electrophile, we identified potent and selective BTK inhibitors that demonstrate biochemical residence times spanning from minutes to 7 days. An inverted cyanoacrylamide with prolonged residence time in vivo remained bound to BTK more than 18 hours after clearance from the circulation. The inverted cyanoacrylamide strategy was further utilized to discover fibroblast growth factor receptor (FGFR) kinase inhibitors with residence times of several days, demonstrating generalizability of the approach. Targeting noncatalytic cysteines with inverted cyanoacrylamides may serve as a broadly applicable platform that facilitates “residence time by design”, the ability to modulate and improve the duration of target engagement in vivo.
Collapse
Affiliation(s)
| | - Jesse M McFarland
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, California, USA
| | - Ville O Paavilainen
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, California, USA
| | | | - Danny Tam
- Principia Biopharma, South San Francisco, California, USA
| | - Vernon T Phan
- Principia Biopharma, South San Francisco, California, USA
| | | | - David Finkle
- Principia Biopharma, South San Francisco, California, USA
| | - Jin Shu
- Principia Biopharma, South San Francisco, California, USA
| | - Vaishali Patel
- Principia Biopharma, South San Francisco, California, USA
| | - Tony Ton
- Principia Biopharma, South San Francisco, California, USA
| | - Xiaoyan Li
- Principia Biopharma, South San Francisco, California, USA
| | | | - Philip A Nunn
- Principia Biopharma, South San Francisco, California, USA
| | - Dane E Karr
- Principia Biopharma, South San Francisco, California, USA
| | | | | | | | - Erik Verner
- Principia Biopharma, South San Francisco, California, USA
| | - Ken A Brameld
- Principia Biopharma, South San Francisco, California, USA
| | - Ronald J Hill
- Principia Biopharma, South San Francisco, California, USA
| | | | - Jack Taunton
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, California, USA
| |
Collapse
|
48
|
Design strategies to address kinetics of drug binding and residence time. Bioorg Med Chem Lett 2015; 25:2019-27. [DOI: 10.1016/j.bmcl.2015.02.027] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 02/06/2015] [Accepted: 02/11/2015] [Indexed: 02/06/2023]
|
49
|
Swinney DC, Beavis P, Chuang KT, Zheng Y, Lee I, Gee P, Deval J, Rotstein DM, Dioszegi M, Ravendran P, Zhang J, Sankuratri S, Kondru R, Vauquelin G. A study of the molecular mechanism of binding kinetics and long residence times of human CCR5 receptor small molecule allosteric ligands. Br J Pharmacol 2015; 171:3364-75. [PMID: 24628038 DOI: 10.1111/bph.12683] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Revised: 02/04/2014] [Accepted: 02/26/2014] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND PURPOSE The human CCR5 receptor is a co-receptor for HIV-1 infection and a target for anti-viral therapy. A greater understanding of the binding kinetics of small molecule allosteric ligand interactions with CCR5 will lead to a better understanding of the binding process and may help discover new molecules that avoid resistance. EXPERIMENTAL APPROACH Using [(3) H] maraviroc as a radioligand, a number of different binding protocols were employed in conjunction with simulations to determine rate constants, kinetic mechanism and mutant kinetic fingerprints for wild-type and mutant human CCR5 with maraviroc, aplaviroc and vicriviroc. KEY RESULTS Kinetic characterization of maraviroc binding to the wild-type CCR5 was consistent with a two-step kinetic mechanism that involved an initial receptor-ligand complex (RA), which transitioned to a more stable complex, R'A, with at least a 13-fold increase in affinity. The dissociation rate from R'A, k-2 , was 1.2 × 10(-3) min(-1) . The maraviroc time-dependent transition was influenced by F85L, W86A, Y108A, I198A and Y251A mutations of CCR5. CONCLUSIONS AND IMPLICATIONS The interaction between maraviroc and CCR5 proceeded according to a multi-step kinetic mechanism, whereby initial mass action binding and later reorganizations of the initial maraviroc-receptor complex lead to a complex with longer residence time. Site-directed mutagenesis identified a kinetic fingerprint of residues that affected the binding kinetics, leading to the conclusion that allosteric ligand binding to CCR5 involved the rearrangement of the binding site in a manner specific to each allosteric ligand.
Collapse
Affiliation(s)
- David C Swinney
- Roche Palo Alto, Palo Alto, CA, USA; Institute for Rare and Neglected Diseases Drug Discovery, Mountain View, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Augmentation of clozapine with amisulpride: an effective therapeutic strategy for violent treatment-resistant schizophrenia patients in a UK high-security hospital. CNS Spectr 2014; 19:403-10. [PMID: 24284256 DOI: 10.1017/s1092852913000874] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Clozapine is used in the management of treatment-resistant schizophrenia and is effective in reducing aggression; however a subgroup of patients is poorly responsive. For violent patients in this group, there is limited literature on the use of strategies to augment clozapine with other agents. Here we present a case series of 6 schizophrenia patients, within a high-security hospital, who have a history of serious violence and who were treated with clozapine augmented with amisulpride. METHODS We reviewed case notes and health records for evidence of violence/aggression and positive factors such as engagement in activities, and Clinical Global Impression (CGI) scores were formulated. We also examined metabolic parameters before and after augmentation. RESULTS All 6 of the patients showed clinical improvement in symptoms and a reduction in their risk of violence to others. Five patients had a reduction in number of violent/aggressive incidents, and all patients showed improvement in engagement in occupational, vocational, and/or psychological work. Metabolic parameters were largely unchanged except for 1 patient whose Body Mass Index (BMI) increased. Five patients reported side effects as unchanged or improved. CONCLUSION These schizophrenia patients with a history of violence showed clinical improvement and reduced aggression and violence with amisulpride augmentation of clozapine. To our knowledge, this is the first report of an antiaggressive benefit of this combination in forensic psychiatric patients. Further studies are warranted to establish the efficacy and anti-aggressive effects of amisulpride augmentation of clozapine.
Collapse
|