1
|
Carr D, Chin C, Chacon T, Herawi MK, Gonzalez M, West R, Morisseau C, Hammock BD, Pecic S, Kandasamy R. 4-Phenyl-thiazole-based dual inhibitors of fatty acid amide hydrolase and soluble epoxide hydrolase do not alleviate orofacial inflammatory pain in female rats. BBA ADVANCES 2024; 6:100119. [PMID: 39246819 PMCID: PMC11379592 DOI: 10.1016/j.bbadva.2024.100119] [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: 05/03/2024] [Revised: 06/20/2024] [Accepted: 08/04/2024] [Indexed: 09/10/2024] Open
Abstract
Pain arising from trigeminal systems such as headache is common, debilitating, and current treatments (e.g., sumatriptan) are limited. New treatments that target novel mechanisms of action may be required to innovate both short- and long-term pain therapy. Fatty acid amide hydrolase and soluble epoxide hydrolase are two pain-related enzymes that regulate pain and inflammation via independent pathways. We have previously demonstrated that simultaneous inhibition of these enzymes using a novel dual inhibitor alleviates acute inflammatory pain in the hindpaw and does not depress wheel running in rats. Here, we expanded on these findings and performed structure-activity relationships of our lead compound, the 4-phenyl-thiazole-based dual inhibitor SW-17, to generate 18 analogs and tested them for their inhibition at both enzymes. Conversion of the sulfonamide group to a tertiary amine led to a general decrease in the potency for the sEH enzyme, while this change was well-tolerated at the FAAH enzyme yielding several strong inhibitors. Six selected inhibitors were evaluated in mouse and rat sEH inhibition assays and results showed a species difference, i.e. 4-phenyl-thiazole-based analogs are significantly less or not active in mouse sEH compared to human and rat enzymes. The most potent inhibitor, SW-17, was evaluated in a plasma stability assay in human and rat plasma and showed moderate stability. However, SW-17 did not alleviate orofacial inflammatory pain in female rats compared to the traditional anti-migraine agent sumatriptan. Although modification of 4-phenyl-thiazole-based dual inhibitor SW-17 changes potencies at both FAAH and sEH, these approaches may not produce antinociception against trigeminal pain. Key Words: polypharmacology, formalin, inflammation, enzyme inhibition, structure-activity relationship studies.
Collapse
Affiliation(s)
- Daniel Carr
- Department of Psychology, California State University, East Bay, Hayward, CA, USA
| | - Christopher Chin
- Department of Psychology, California State University, East Bay, Hayward, CA, USA
| | - Tiffany Chacon
- Department of Psychology, California State University, East Bay, Hayward, CA, USA
| | - Monijeh Khoja Herawi
- Department of Psychology, California State University, East Bay, Hayward, CA, USA
| | - Michael Gonzalez
- Department of Chemistry and Biochemistry, California State University, Fullerton, Fullerton, CA, USA
| | - Ryan West
- Department of Chemistry and Biochemistry, California State University, Fullerton, Fullerton, CA, USA
| | - Christophe Morisseau
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, Davis, CA, USA
| | - Bruce D Hammock
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, Davis, CA, USA
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University, Fullerton, Fullerton, CA, USA
| | - Ram Kandasamy
- Department of Psychology, California State University, East Bay, Hayward, CA, USA
| |
Collapse
|
2
|
Dey S, Biradar R, Mane SS, Kunnath Shaji A, Das AP, Agarwal SM, Dengale SJ. Identification and characterization of the in-vivo metabolites of the novel soluble epoxide hydrolase inhibitor EC5026 using liquid chromatography quadrupole time of flight mass spectrometry. J Pharm Biomed Anal 2024; 244:116116. [PMID: 38537542 DOI: 10.1016/j.jpba.2024.116116] [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: 12/30/2023] [Revised: 02/28/2024] [Accepted: 03/15/2024] [Indexed: 04/29/2024]
Abstract
EC5026 is a novel soluble epoxide hydrolase inhibitor being developed clinically to treat neuropathic pain and inflammation. In the current study, we employed the LC-ESI-Q-TOF-MS/MS technique to identify four in-vivo phase-I metabolites of EC5026 in rat model, out of which three were found to be novel. The identified metabolites include aliphatic hydroxylation, di-hydroxylation, terminal desaturation, and carboxylation. No phase-II metabolites were found. The pharmacokinetic profile of identified metabolites was established after a single oral dose of EC5026 to Wistar rats. The Tmax of the drug and metabolites were found to be in the range of 1-2 hours and 4-12 hours, respectively. The major metabolites M1 and M2 were found to have more than 2-fold (263.87% AUC) and equivalent exposure (96.33% AUC) compared to the parent drug, respectively. Further, the docking study revealed that the mono-hydroxylated and terminally desaturated metabolites possess better binding affinity than the parent drug. Therefore, these metabolites may hold sEH inhibition potential and can be followed through future research.
Collapse
Affiliation(s)
- Shankha Dey
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Changsari 781101, India
| | - Rushikesh Biradar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Changsari 781101, India
| | - Sayalee Sanjay Mane
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Changsari 781101, India
| | - Anandhu Kunnath Shaji
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Changsari 781101, India
| | - Agneesh Pratim Das
- ICMR-National Institute of Cancer Prevention and Research, I-7, Sector-39, Noida 201301, India
| | - Subhash Mohan Agarwal
- ICMR-National Institute of Cancer Prevention and Research, I-7, Sector-39, Noida 201301, India
| | - Swapnil Jayant Dengale
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Changsari 781101, India.
| |
Collapse
|
3
|
Bu Y, Yang S, Wang D, Hu S, Zhang Q, Wu Z, Yang C. Role of soluble epoxide hydrolase in pain and depression comorbidity. Neurobiol Dis 2024; 193:106443. [PMID: 38395315 DOI: 10.1016/j.nbd.2024.106443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 02/25/2024] Open
Abstract
The coexistence of chronic pain and depression in clinical practice places a substantial social burden and profoundly impacts in patients. Although a clear correlation exists, the underlying mechanism of comorbidity between chronic pain and depression remains elusive. Research conducted in recent decades has uncovered that soluble epoxide hydrolase, a pivotal enzyme in the metabolism of polyunsaturated fatty acids, plays a crucial role in inflammation. Interestingly, this enzyme is intricately linked to the development of both pain and depression. With this understanding, this review aims to summarize the roles of soluble epoxide hydrolase in pain, depression, and their comorbidity. Simultaneously, we will also explore the underlying mechanisms, providing guidance for future research and drug development.
Collapse
Affiliation(s)
- Yuchen Bu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Siqi Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Di Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Suwan Hu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qi Zhang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zifeng Wu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
4
|
Biradar R, Dey S, Mane SS, Dengale SJ. Development and validation of LC-MS/MS method for estimating the pharmacokinetics, protein binding, and metabolic stability of soluble epoxide hydrolase inhibitor EC5026. J Pharm Biomed Anal 2024; 237:115797. [PMID: 37847987 DOI: 10.1016/j.jpba.2023.115797] [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: 05/21/2023] [Revised: 07/17/2023] [Accepted: 10/11/2023] [Indexed: 10/19/2023]
Abstract
EC5026 is a soluble epoxide hydrolase (SEH) inhibitor which is being developed clinically (Phase-1) as a first-in-class analgesic for the treatment of pain. In the present study, we report the development and validation of the LC-MS/MS method for the estimation of EC5026 from rat plasma. The developed method is simple, specific, and sensitive for the quantification of EC5026 from rat plasma. The method was applied to investigate in-vivo pharmacokinetics after single oral administration (P.O.) of EC5026 (5.0 mg/kg) in Wistar rats. Further, the in-vitro metabolic stability and rat plasma protein binding of EC5026 were evaluated using the developed method. The in-vitro results demonstrated a moderate clearance with a value of 10.8 mL/min/kg and half-life (t1/2) of 57.75 min. The results show moderate clearance by the liver manifesting in satisfactory oral bioavailability. The rat plasma protein binding was estimated to be 96.24% ± 0.97% and 96.38% ± 0.56% at 1 µM and 10 µM concentrations, respectively. The developed analytical method is expected to facilitate future pre-clinical, clinical investigations of EC5026.
Collapse
Affiliation(s)
- Rushikesh Biradar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Changsari, Guwahati 781 101, India
| | - Shankha Dey
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Changsari, Guwahati 781 101, India
| | - Sayalee Sanjay Mane
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Changsari, Guwahati 781 101, India
| | - Swapnil J Dengale
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Changsari, Guwahati 781 101, India.
| |
Collapse
|
5
|
Prasher P, Mall T, Sharma M. Synthesis and biological profile of benzoxazolone derivatives. Arch Pharm (Weinheim) 2023; 356:e2300245. [PMID: 37379239 DOI: 10.1002/ardp.202300245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/30/2023]
Abstract
The benzoxazolone nucleus is an ideal scaffold for drug design, owing to its discrete physicochemical profile, bioisosteric preference over pharmacokinetically weaker moieties, weakly acidic behavior, presence of both lipophilic and hydrophilic fragments on a single framework, and a wider choice of chemical modification on the benzene and oxazolone rings. These properties apparently influence the interactions of benzoxazolone-based derivatives with their respective biological targets. Hence, the benzoxazolone ring is implicated in the synthesis and development of pharmaceuticals with a diverse biological profile ranging from anticancer, analgesics, insecticides, anti-inflammatory, and neuroprotective agents. This has further led to the commercialization of several benzoxazolone-based molecules and a few others under clinical trials. Nevertheless, the SAR exploration of benzoxazolone derivatives for the identification of potential "hits" followed by the screening of "leads" provides a plethora of opportunities for further exploration of the pharmacological profile of the benzoxazolone nucleus. In this review, we aim to present the biological profile of different derivatives based on the benzoxazolone framework.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, India
| | - Tanisqa Mall
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, India
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Dehradun, India
| |
Collapse
|
6
|
Takeshita AA, Hammock BD, Wagner KM. Soluble epoxide hydrolase inhibition alleviates chemotherapy induced neuropathic pain. FRONTIERS IN PAIN RESEARCH (LAUSANNE, SWITZERLAND) 2023; 3:1100524. [PMID: 36700145 PMCID: PMC9868926 DOI: 10.3389/fpain.2022.1100524] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 12/15/2022] [Indexed: 01/12/2023]
Abstract
Chemotherapy induced peripheral neuropathy (CIPN) is a particularly pernicious form of neuropathy and the associated pain is the primary dose-limiting factor of life-prolonging chemotherapy treatment. The prevalence of CIPN is high and can last long after treatment has been stopped. Currently, late in the COVID-19 pandemic, there are still increased psychological pressures on cancer patients as well as additional challenges in providing analgesia for them. These include the risks of nonsteroidal anti-inflammatory drug (NSAID) analgesics potentially masking early infection symptoms and the immunosuppression of steroidal and opiate based approaches. Even without these concerns, CIPN is often inadequately treated with few therapies that offer significant pain relief. The experiments we report use soluble epoxide hydrolase inhibitors (sEHI) which relieved this intractable pain in preclinical models. Doses of EC5026, an IND candidate intended to treat neuropathic pain, elicited dose dependent analgesic responses in multiple models including platinum-based, taxane, and vinca alkaloid-based CIPN pain in Sprague Dawley rats. At the same time as a class, the sEHI are known to result in fewer debilitating side effects of other analgesics, likely due to their novel mechanism of action. Overall, the observed dose-dependent analgesia in both male and female rats across multiple models of chemotherapy induced neuropathic pain holds promise as a useful tool when translated to the clinic.
Collapse
Affiliation(s)
| | - Bruce D. Hammock
- EicOsis LLC, Davis, CA, United States,Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, United States
| | - Karen M. Wagner
- EicOsis LLC, Davis, CA, United States,Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, United States,Correspondence: Karen M. Wagner ;
| |
Collapse
|
7
|
Lipid mediators generated by the cytochrome P450—Epoxide hydrolase pathway. ADVANCES IN PHARMACOLOGY 2023; 97:327-373. [DOI: 10.1016/bs.apha.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
8
|
Codony S, Entrena JM, Calvó-Tusell C, Jora B, González-Cano R, Osuna S, Corpas R, Morisseau C, Pérez B, Barniol-Xicota M, Griñán-Ferré C, Pérez C, Rodríguez-Franco MI, Martínez AL, Loza MI, Pallàs M, Verhelst SHL, Sanfeliu C, Feixas F, Hammock BD, Brea J, Cobos EJ, Vázquez S. Synthesis, In Vitro Profiling, and In Vivo Evaluation of Benzohomoadamantane-Based Ureas for Visceral Pain: A New Indication for Soluble Epoxide Hydrolase Inhibitors. J Med Chem 2022; 65:13660-13680. [PMID: 36222708 PMCID: PMC9620236 DOI: 10.1021/acs.jmedchem.2c00515] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The soluble epoxide hydrolase (sEH) has been suggested as a pharmacological target for the treatment of several diseases, including pain-related disorders. Herein, we report further medicinal chemistry around new benzohomoadamantane-based sEH inhibitors (sEHI) in order to improve the drug metabolism and pharmacokinetics properties of a previous hit. After an extensive in vitro screening cascade, molecular modeling, and in vivo pharmacokinetics studies, two candidates were evaluated in vivo in a murine model of capsaicin-induced allodynia. The two compounds showed an anti-allodynic effect in a dose-dependent manner. Moreover, the most potent compound presented robust analgesic efficacy in the cyclophosphamide-induced murine model of cystitis, a well-established model of visceral pain. Overall, these results suggest painful bladder syndrome as a new possible indication for sEHI, opening a new range of applications for them in the visceral pain field.
Collapse
Affiliation(s)
- Sandra Codony
- Laboratori
de Química Farmacèutica (Unitat Associada al CSIC),
Facultat de Farmàcia i Ciències de l’Alimentació,
and Institute of Biomedicine (IBUB), Universitat
de Barcelona, Av. Joan XXIII, 27-31, Barcelona 08028, Spain
| | - José M. Entrena
- Animal
Behavior Research Unit, Scientific Instrumentation Center, Parque
Tecnológico de Ciencias de la Salud, University of Granada, Armilla, Granada 18100, Spain
| | - Carla Calvó-Tusell
- CompBioLab
Group, Departament de Química and Institut de Química
Computacional i Catàlisi (IQCC), Universitat de Girona, C/ Maria Aurèlia Capmany 69, Girona 17003, Spain
| | - Beatrice Jora
- Laboratori
de Química Farmacèutica (Unitat Associada al CSIC),
Facultat de Farmàcia i Ciències de l’Alimentació,
and Institute of Biomedicine (IBUB), Universitat
de Barcelona, Av. Joan XXIII, 27-31, Barcelona 08028, Spain
| | - Rafael González-Cano
- Department
of Pharmacology, Faculty of Medicine and Biomedical Research Center
(Neurosciences Institute), Biosanitary Research Institute ibs.GRANADA, University of Granada, Avenida de la Investigación 11, Granada 18016, Spain
| | - Sílvia Osuna
- CompBioLab
Group, Departament de Química and Institut de Química
Computacional i Catàlisi (IQCC), Universitat de Girona, C/ Maria Aurèlia Capmany 69, Girona 17003, Spain,Institució
Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain
| | - Rubén Corpas
- Institute
of Biomedical Research of Barcelona (IIBB), CSIC and IDIBAPS, Barcelona 08036, Spain
| | - Christophe Morisseau
- Department
of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Belén Pérez
- Department
of Pharmacology, Therapeutics and Toxicology, Institute of Neurosciences, Autonomous University of Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Marta Barniol-Xicota
- Laboratory
of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven—University of Leuven, Herestraat 49 box B901, Leuven 3000, Belgium
| | - Christian Griñán-Ferré
- Pharmacology
Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry,
Faculty of Pharmacy and Food Sciences, Institute of Neuroscience, University of Barcelona (NeuroUB), Av. Joan XXIII 27-31, Barcelona 08028, Spain
| | - Concepción Pérez
- Institute of Medicinal Chemistry, Spanish
National Research Council (CSIC), C/Juan de la Cierva 3, Madrid 28006, Spain
| | - María Isabel Rodríguez-Franco
- Institute of Medicinal Chemistry, Spanish
National Research Council (CSIC), C/Juan de la Cierva 3, Madrid 28006, Spain
| | - Antón L. Martínez
- Drug Screening
Platform/Biofarma Research Group, CIMUS Research Center, University of Santiago de Compostela (USC), Santiago de Compostela 15782, Spain
| | - M. Isabel Loza
- Drug Screening
Platform/Biofarma Research Group, CIMUS Research Center, University of Santiago de Compostela (USC), Santiago de Compostela 15782, Spain
| | - Mercè Pallàs
- Pharmacology
Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry,
Faculty of Pharmacy and Food Sciences, Institute of Neuroscience, University of Barcelona (NeuroUB), Av. Joan XXIII 27-31, Barcelona 08028, Spain
| | - Steven H. L. Verhelst
- Laboratory
of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven—University of Leuven, Herestraat 49 box B901, Leuven 3000, Belgium,Leibniz Institute
for Analytical Sciences ISAS, AG Chemical
Proteomics, Otto-Hahn-Str.
6b, Dortmund 44227, Germany
| | - Coral Sanfeliu
- Institute
of Biomedical Research of Barcelona (IIBB), CSIC and IDIBAPS, Barcelona 08036, Spain
| | - Ferran Feixas
- CompBioLab
Group, Departament de Química and Institut de Química
Computacional i Catàlisi (IQCC), Universitat de Girona, C/ Maria Aurèlia Capmany 69, Girona 17003, Spain
| | - Bruce D. Hammock
- Department
of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - José Brea
- Drug Screening
Platform/Biofarma Research Group, CIMUS Research Center, University of Santiago de Compostela (USC), Santiago de Compostela 15782, Spain
| | - Enrique J. Cobos
- Department
of Pharmacology, Faculty of Medicine and Biomedical Research Center
(Neurosciences Institute), Biosanitary Research Institute ibs.GRANADA, University of Granada, Avenida de la Investigación 11, Granada 18016, Spain
| | - Santiago Vázquez
- Laboratori
de Química Farmacèutica (Unitat Associada al CSIC),
Facultat de Farmàcia i Ciències de l’Alimentació,
and Institute of Biomedicine (IBUB), Universitat
de Barcelona, Av. Joan XXIII, 27-31, Barcelona 08028, Spain,. Phone: +34 934024533
| |
Collapse
|
9
|
Kwatra SG, Misery L, Clibborn C, Steinhoff M. Molecular and cellular mechanisms of itch and pain in atopic dermatitis and implications for novel therapeutics. Clin Transl Immunology 2022; 11:e1390. [PMID: 35582626 PMCID: PMC9082890 DOI: 10.1002/cti2.1390] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 12/03/2022] Open
Abstract
Atopic dermatitis is a chronic inflammatory skin disease. Patients with atopic dermatitis experience inflammatory lesions associated with intense itch and pain, which lead to sleep disturbance and poor mental health and quality of life. We review the molecular mechanisms underlying itch and pain symptoms in atopic dermatitis and discuss the current clinical development of treatments for moderate-to-severe atopic dermatitis. The molecular pathology of atopic dermatitis includes aberrant immune activation involving significant cross-talk among the skin and immune and neuronal cells. Exogenous and endogenous triggers modulate stimulation of mediators including cytokine/chemokine expression/release by the skin and immune cells, which causes inflammation, skin barrier disruption, activation and growth of sensory neurons, itch and pain. These complex interactions among cell types are mediated primarily by cytokines, but also involve chemokines, neurotransmitters, lipids, proteases, antimicrobial peptides, agonists of ion channels or various G protein-coupled receptors. Patients with atopic dermatitis have a cytokine profile characterised by abnormal levels of interleukins 4, 12, 13, 18, 22, 31 and 33; thymic stromal lymphopoietin; and interferon gamma. Cytokine receptors mainly signal through the Janus kinase/signal transducer and activator of transcription pathway. Among emerging novel therapeutics, several Janus kinase inhibitors are being developed for topical or systemic treatment of moderate-to-severe atopic dermatitis because of their potential to modulate cytokine expression and release. Janus kinase inhibitors lead to changes in gene expression that have favourable effects on local and systemic cytokine release, and probably other mediators, thus successfully modulating molecular mechanisms responsible for itch and pain in atopic dermatitis.
Collapse
Affiliation(s)
- Shawn G Kwatra
- Department of DermatologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Laurent Misery
- Department of DermatologyUniversity Hospital of BrestBrestFrance
| | | | - Martin Steinhoff
- Department of Dermatology and VenereologyHamad Medical CorporationDohaQatar
- Translational Research InstituteAcademic Health SystemHamad Medical CorporationDohaQatar
- Dermatology InstituteAcademic Health SystemHamad Medical CorporationDohaQatar
- Department of DermatologyWeill Cornell Medicine‐QatarDohaQatar
- Qatar University, College of MedicineDohaQatar
- Department of DermatologyWeill Cornell MedicineNew YorkNYUSA
| |
Collapse
|
10
|
Du F, Cao R, Chen L, Sun J, Shi Y, Fu Y, Hammock BD, Zheng Z, Liu Z, Chen G. Structure-guided discovery of potent and oral soluble epoxide hydrolase inhibitors for the treatment of neuropathic pain. Acta Pharm Sin B 2022; 12:1377-1389. [PMID: 35530144 PMCID: PMC9072249 DOI: 10.1016/j.apsb.2021.09.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 11/01/2022] Open
Abstract
Soluble epoxide hydrolase (sEH) is related to arachidonic acid cascade and is over-expressed in a variety of diseases, making sEH an attractive target for the treatment of pain as well as inflammatory-related diseases. A new series of memantyl urea derivatives as potent sEH inhibitors was obtained using our previous reported compound 4 as lead compound. A preferential modification of piperidinyl to 3-carbamoyl piperidinyl was identified for this series via structure-based rational drug design. Compound A20 exhibited moderate percentage plasma protein binding (88.6%) and better metabolic stability in vitro. After oral administration, the bioavailability of A20 was 28.6%. Acute toxicity test showed that A20 was well tolerated and there was no adverse event encountered at dose of 6.0 g/kg. Inhibitor A20 also displayed robust analgesic effect in vivo and dose-dependently attenuated neuropathic pain in rat model induced by spared nerve injury, which was better than gabapentin and sEH inhibitor (±)-EC-5026. In one word, the oral administration of A20 significantly alleviated pain and improved the health status of the rats, demonstrating that A20 was a promising candidate to be further evaluated for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Fangyu Du
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ruolin Cao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lu Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jianwen Sun
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yajie Shi
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yang Fu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bruce D. Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, USA
| | - Zhonghui Zheng
- Shandong Xinhua Pharmaceutical Co., Ltd., Zibo 255086, China
| | - Zhongbo Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Guoliang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
11
|
Du F, Sun W, Morisseau C, Hammock BD, Bao X, Liu Q, Wang C, Zhang T, Yang H, Zhou J, Xiao W, Liu Z, Chen G. Discovery of memantyl urea derivatives as potent soluble epoxide hydrolase inhibitors against lipopolysaccharide-induced sepsis. Eur J Med Chem 2021; 223:113678. [PMID: 34218083 PMCID: PMC8877601 DOI: 10.1016/j.ejmech.2021.113678] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 06/18/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022]
Abstract
Sepsis, a systemic inflammatory response, caused by pathogenic factors including microorganisms, has high mortality and limited therapeutic approaches. Herein, a new soluble epoxide hydrolase (sEH) inhibitor series comprising a phenyl ring connected to a memantyl moiety via a urea or amide linkage has been designed. A preferential urea pharmacophore that improved the binding properties of the compounds was identified for those series via biochemical assay in vitro and in vivo studies. Molecular docking displayed that 3,5-dimethyl on the adamantyl group in B401 could make van der Waals interactions with residues at a hydrophobic pocket of sEH active site, which might indirectly explain the subnanomolar level activities of memantyl urea derivatives in vitro better than AR-9281. Among them, compound B401 significantly improved the inhibition potency with human and murine sEH IC50 values as 0.4 nM and 0.5 nM, respectively. Although the median survival time of C57BL/6 mice in LPS-induced sepsis model was slightly increased, the survival rate did not reach significant efficacy. Based on safety profile, metabolic stability, pharmacokinetic and in vivo efficacy, B401 demonstrated the proof of potential for this class of memantyl urea-based sEH inhibitors as therapeutic agents in sepsis.
Collapse
Affiliation(s)
- Fangyu Du
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, China
| | - Wenjiao Sun
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, China
| | - Christophe Morisseau
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Bruce D Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Xuefei Bao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, China; Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang, Jiangsu, 222001, China
| | - Qiu Liu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang, Jiangsu, 222001, China
| | - Chao Wang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang, Jiangsu, 222001, China
| | - Tan Zhang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang, Jiangsu, 222001, China
| | - Hao Yang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang, Jiangsu, 222001, China
| | - Jun Zhou
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang, Jiangsu, 222001, China
| | - Wei Xiao
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang, Jiangsu, 222001, China.
| | - Zhongbo Liu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, China.
| | - Guoliang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
12
|
Wang J, Fang Y, Fan RA, Kirk CJ. Proteasome Inhibitors and Their Pharmacokinetics, Pharmacodynamics, and Metabolism. Int J Mol Sci 2021; 22:ijms222111595. [PMID: 34769030 PMCID: PMC8583966 DOI: 10.3390/ijms222111595] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 12/15/2022] Open
Abstract
The proteasome is responsible for mediating intracellular protein degradation and regulating cellular function with impact on tumor and immune effector cell biology. The proteasome is found predominantly in two forms, the constitutive proteasome and the immunoproteasome. It has been validated as a therapeutic drug target through regulatory approval with 2 distinct chemical classes of small molecular inhibitors (boronic acid derivatives and peptide epoxyketones), including 3 compounds, bortezomib (VELCADE), carfilzomib (KYPROLIS), and ixazomib (NINLARO), for use in the treatment of the plasma cell neoplasm, multiple myeloma. Additionally, a selective inhibitor of immunoproteasome (KZR-616) is being developed for the treatment of autoimmune diseases. Here, we compare and contrast the pharmacokinetics (PK), pharmacodynamics (PD), and metabolism of these 2 classes of compounds in preclinical models and clinical studies. The distinct metabolism of peptide epoxyketones, which is primarily mediated by microsomal epoxide hydrolase, is highlighted and postulated as a favorable property for the development of this class of compound in chronic conditions.
Collapse
|
13
|
Iadarola MJ, Brown DC, Nahama A, Sapio MR, Mannes AJ. Pain Treatment in the Companion Canine Model to Validate Rodent Results and Incentivize the Transition to Human Clinical Trials. Front Pharmacol 2021; 12:705743. [PMID: 34421597 PMCID: PMC8375595 DOI: 10.3389/fphar.2021.705743] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/14/2021] [Indexed: 12/14/2022] Open
Abstract
One of the biggest challenges for analgesic drug development is how to decide if a potential analgesic candidate will work in humans. What preclinical data are the most convincing, incentivizing and most predictive of success? Such a predicament is not unique to analgesics, and the pain field has certain advantages over drug development efforts in areas like neuropsychiatry where the etiological origins are either unknown or difficult to ascertain. For pain, the origin of the problem frequently is known, and the causative peripheral tissue insult might be observable. The main conundrum centers around evaluation of translational cell- and rodent-based results. While cell and rodent models are undeniably important first steps for screening, probing mechanism of action, and understanding factors of adsorption, distribution metabolism and excretion, two questions arise from such studies. First, are they reliable indicators of analgesic performance of a candidate drug in human acute and chronic pain? Second, what additional model systems might be capable of increasing translational confidence? We address this second question by assessing, primarily, the companion canine model, which can provide particularly strong predictive information for candidate analgesic agents in humans. This statement is mainly derived from our studies with resiniferatoxin (RTX) a potent TRPV1 agonist but also from protein therapeutics using a conjugate of Substance P and saporin. Our experience, to date, is that rodent models might be very well suited for acute pain translation, but companion canine models, and other large animal studies, can augment initial discovery research using rodent models for neuropathic or chronic pain. The larger animal models also provide strong translational predictive capacity for analgesic performance in humans, better predict dosing parameters for human trials and provide insight into behavior changes (bladder, bowel, mood, etc.) that are not readily assessed in laboratory animals. They are, however, not without problems that can be encountered with any experimental drug treatment or clinical trial. It also is important to recognize that pain treatment is a major veterinary concern and is an intrinsically worthwhile endeavor for animals as well as humans.
Collapse
Affiliation(s)
- Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, NIH, Bethesda, MD, United States
| | | | | | - Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, NIH, Bethesda, MD, United States
| | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, NIH, Bethesda, MD, United States
| |
Collapse
|
14
|
Jonnalagadda D, Wan D, Chun J, Hammock BD, Kihara Y. A Soluble Epoxide Hydrolase Inhibitor, 1-TrifluoromethoxyPhenyl-3-(1-Propionylpiperidin-4-yl) Urea, Ameliorates Experimental Autoimmune Encephalomyelitis. Int J Mol Sci 2021; 22:ijms22094650. [PMID: 33925035 PMCID: PMC8125305 DOI: 10.3390/ijms22094650] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Polyunsaturated fatty acids (PUFAs) are essential FAs for human health. Cytochrome P450 oxygenates PUFAs to produce anti-inflammatory and pain-resolving epoxy fatty acids (EpFAs) and other oxylipins whose epoxide ring is opened by the soluble epoxide hydrolase (sEH/Ephx2), resulting in the formation of toxic and pro-inflammatory vicinal diols (dihydroxy-FAs). Pharmacological inhibition of sEH is a promising strategy for the treatment of pain, inflammation, cardiovascular diseases, and other conditions. We tested the efficacy of a potent, selective sEH inhibitor, 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU), in an animal model of multiple sclerosis (MS), experimental autoimmune encephalomyelitis (EAE). Prophylactic TPPU treatment significantly ameliorated EAE without affecting circulating white blood cell counts. TPPU accumulated in the spinal cords (SCs), which was correlated with plasma TPPU concentration. Targeted lipidomics in EAE SCs and plasma identified that TPPU blocked production of dihydroxy-FAs efficiently and increased some EpFA species including 12(13)-epoxy-octadecenoic acid (12(13)-EpOME) and 17(18)-epoxy-eicosatrienoic acid (17(18)-EpETE). TPPU did not alter levels of cyclooxygenase (COX-1/2) metabolites, while it increased 12-hydroxyeicosatetraenoic acid (12-HETE) and other 12/15-lipoxygenase metabolites. These analytical results are consistent with sEH inhibitors that reduce neuroinflammation and accelerate anti-inflammatory responses, providing the possibility that sEH inhibitors could be used as a disease modifying therapy, as well as for MS-associated pain relief.
Collapse
Affiliation(s)
- Deepa Jonnalagadda
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA; (D.J.); (J.C.)
| | - Debin Wan
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA 95817, USA; (D.W.); (B.D.H.)
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA; (D.J.); (J.C.)
| | - Bruce D. Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA 95817, USA; (D.W.); (B.D.H.)
| | - Yasuyuki Kihara
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA; (D.J.); (J.C.)
- Correspondence:
| |
Collapse
|
15
|
Hammock B, McReynolds CB, Wagner K, Buckpitt A, Cortes-Puch I, Croston G, Lee KSS, Yang J, Schmidt WK, Hwang SH. Movement to the Clinic of Soluble Epoxide Hydrolase Inhibitor EC5026 as an Analgesic for Neuropathic Pain and for Use as a Nonaddictive Opioid Alternative. J Med Chem 2021; 64:1856-1872. [PMID: 33550801 PMCID: PMC7917437 DOI: 10.1021/acs.jmedchem.0c01886] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Indexed: 12/12/2022]
Abstract
This report describes the development of an orally active analgesic that resolves inflammation and neuropathic pain without the addictive potential of opioids. EC5026 acts on the cytochrome P450 branch of the arachidonate cascade to stabilize epoxides of polyunsaturated fatty acids (EpFA), which are natural mediators that reduce pain, resolve inflammation, and maintain normal blood pressure. EC5026 is a slow-tight binding transition-state mimic that inhibits the soluble epoxide hydrolase (sEH) at picomolar concentrations. The sEH rapidly degrades EpFA; thus, inhibiting sEH increases EpFA in vivo and confers beneficial effects. This mechanism addresses disease states by shifting endoplasmic reticulum stress from promoting cellular senescence and inflammation toward cell survival and homeostasis. We describe the synthesis and optimization of EC5026 and its development through human Phase 1a trials with no drug-related adverse events. Additionally, we outline fundamental work leading to discovery of the analgesic and inflammation-resolving CYP450 branch of the arachidonate cascade.
Collapse
Affiliation(s)
- Bruce
D. Hammock
- EicOsis
Human Health Inc., Subsidiary of EicOsis LLC, 1930 Fifth Street, Suite A, Davis, California 95616, United States
| | - Cindy B. McReynolds
- EicOsis
Human Health Inc., Subsidiary of EicOsis LLC, 1930 Fifth Street, Suite A, Davis, California 95616, United States
| | - Karen Wagner
- EicOsis
Human Health Inc., Subsidiary of EicOsis LLC, 1930 Fifth Street, Suite A, Davis, California 95616, United States
| | - Alan Buckpitt
- EicOsis
Human Health Inc., Subsidiary of EicOsis LLC, 1930 Fifth Street, Suite A, Davis, California 95616, United States
| | - Irene Cortes-Puch
- EicOsis
Human Health Inc., Subsidiary of EicOsis LLC, 1930 Fifth Street, Suite A, Davis, California 95616, United States
| | - Glenn Croston
- EicOsis
Human Health Inc., Subsidiary of EicOsis LLC, 1930 Fifth Street, Suite A, Davis, California 95616, United States
| | | | - Jun Yang
- EicOsis
Human Health Inc., Subsidiary of EicOsis LLC, 1930 Fifth Street, Suite A, Davis, California 95616, United States
| | - William K. Schmidt
- EicOsis
Human Health Inc., Subsidiary of EicOsis LLC, 1930 Fifth Street, Suite A, Davis, California 95616, United States
| | - Sung Hee Hwang
- EicOsis
Human Health Inc., Subsidiary of EicOsis LLC, 1930 Fifth Street, Suite A, Davis, California 95616, United States
| |
Collapse
|
16
|
Vorobjeva NV, Sud'ina GF, Chernyak BV. Mitochondria Are Potential Targets for the Development of New Drugs Against Neutrophilic Inflammation in Severe Pneumonia Including COVID-19. Front Pharmacol 2021; 12:609508. [PMID: 33584318 PMCID: PMC7878366 DOI: 10.3389/fphar.2021.609508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/04/2021] [Indexed: 12/26/2022] Open
Affiliation(s)
- Nina V Vorobjeva
- Biology Faculty, M. V. Lomonosov Moscow State University, Moscow, Russia
| | - Galina F Sud'ina
- A. N. Belozersky Institute of Physico-Chemical Biology, M. V. Lomonosov Moscow State University, Moscow, Russia
| | - Boris V Chernyak
- A. N. Belozersky Institute of Physico-Chemical Biology, M. V. Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
17
|
Ruiz-Cantero MC, González-Cano R, Tejada MÁ, Santos-Caballero M, Perazzoli G, Nieto FR, Cobos EJ. Sigma-1 receptor: A drug target for the modulation of neuroimmune and neuroglial interactions during chronic pain. Pharmacol Res 2021; 163:105339. [PMID: 33276102 DOI: 10.1016/j.phrs.2020.105339] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022]
Abstract
Immune and glial cells play a pivotal role in chronic pain. Therefore, it is possible that the pharmacological modulation of neurotransmission from an exclusively neuronal perspective may not be enough for adequate pain management, and the modulation of complex interactions between neurons and other cell types might be needed for successful pain relief. In this article, we review the current scientific evidence for the modulatory effects of sigma-1 receptors on communication between the immune and nervous systems during inflammation, as well as the influence of this receptor on peripheral and central neuroinflammation. Several experimental models of pathological pain are considered, including peripheral and central neuropathic pain, osteoarthritic, and cancer pain. Sigma-1 receptor inhibition prevents peripheral (macrophage infiltration into the dorsal root ganglion) and central (activation of microglia and astrocytes) neuroinflammation in several pain models, and enhances immune-driven peripheral opioid analgesia during painful inflammation, maximizing the analgesic potential of peripheral immune cells. Therefore, sigma-1 antagonists may constitute a new class of analgesics with an unprecedented mechanism of action and potential utility in several painful disorders.
Collapse
Affiliation(s)
- M Carmen Ruiz-Cantero
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, Granada, Spain; Biosanitary Research Institute ibs.GRANADA, Granada, Spain
| | - Rafael González-Cano
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, Granada, Spain; Biosanitary Research Institute ibs.GRANADA, Granada, Spain
| | - Miguel Á Tejada
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, Granada, Spain; INCLIVA Health Research Institute, Valencia, Spain
| | - Miriam Santos-Caballero
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, Granada, Spain; Biosanitary Research Institute ibs.GRANADA, Granada, Spain
| | - Gloria Perazzoli
- Biosanitary Research Institute ibs.GRANADA, Granada, Spain; Department of Nursing, Physiotherapy and Medicine, University of Almería, Almería, Spain
| | - Francisco R Nieto
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, Granada, Spain; Biosanitary Research Institute ibs.GRANADA, Granada, Spain.
| | - Enrique J Cobos
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, Granada, Spain; Biosanitary Research Institute ibs.GRANADA, Granada, Spain; Teófilo Hernando Institute for Drug Discovery, Madrid, Spain.
| |
Collapse
|
18
|
Sang CN, Schmidt WK. Aligning New Approaches to Accelerate the Development of Non-opioid Analgesic Therapies. Neurotherapeutics 2020; 17:765-769. [PMID: 33058022 PMCID: PMC7609808 DOI: 10.1007/s13311-020-00935-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Affiliation(s)
- Christine N Sang
- Translational Pain Research, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| | | |
Collapse
|