1
|
Shinotsuka N, Shimizu H, Komatsu T, Ito A, Yoshikawa S, Takashima T, Sugiura A, Moriguchi Y, Ohno Y, Yamasawa M, Noguchi H, Fujiuchi A, Konishi K. AK1780, a selective P2X7 receptor antagonist with high central nervous system penetration, exhibits analgesic effect in rat neuropathic pain model. THE JOURNAL OF PAIN 2025; 31:105380. [PMID: 40158710 DOI: 10.1016/j.jpain.2025.105380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/28/2025] [Accepted: 03/16/2025] [Indexed: 04/02/2025]
Abstract
It is widely recognized that the mechanisms underlying neuropathic pain are complicated, and no existing analgesics give sufficient effects to all patients suffering it, as such there remains a high unmet medical need for well tolerated analgesics. The P2X7 receptor (P2X7R), an adenosine triphosphate-activated ion-gated channel, is an attractive therapeutic target for neuropathic pain according to previous in vitro and in vivo studies. Here, we introduce a novel P2X7R inhibitor, AK1780 (LY3857210). AK1780 inhibited calcium influx in human or rat P2X7R expressing cells with IC50 of 19 or 8.4 nmol/L, respectively, while AK1780 did not show activity against other P2X family as well as a non-binding profile for 133 other targets. Furthermore, AK1780 suppressed IL-1β secretion from rat primary microglial cells which may play an important role in pain sensation. AK1780 attenuated paw withdrawal threshold after 2 mg/kg oral administration, in which both plasma and CSF concentrations achieved in vitro IC90, determined by calcium influx assay, with no apparent CNS side effects. More interestingly, the receptor occupancy in the brain was correlated with analgesic effects in rats. These data suggest that blockade of P2X7R with AK1780 in the CNS can have analgesic properties without any CNS adverse effects. PERSPECTIVE: This article presents in vitro and in vivo data for AK1780 (LY3857210), a selective P2X7R antagonist. The data reasonably explain its analgesic effect in the view of PK/PD, indicating that AK1780 may potentially suppress neuropathic pain without any CNS adverse effects.
Collapse
Affiliation(s)
- Naomi Shinotsuka
- Pharmaceuticals Research Centre, Asahi Kasei Pharma Corporation, 632-1, Mifuku, Izunokuni, Shizuoka 410-2321, Japan.
| | - Hirotomo Shimizu
- Pharmaceuticals Research Centre, Asahi Kasei Pharma Corporation, 632-1, Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Takayuki Komatsu
- Pharmaceuticals Research Centre, Asahi Kasei Pharma Corporation, 632-1, Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Akitoshi Ito
- Pharmaceuticals Research Centre, Asahi Kasei Pharma Corporation, 632-1, Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Satoru Yoshikawa
- Pharmaceuticals Research Centre, Asahi Kasei Pharma Corporation, 632-1, Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Tadayuki Takashima
- Pharmaceuticals Research Centre, Asahi Kasei Pharma Corporation, 632-1, Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Akemi Sugiura
- RaQualia Pharma Inc., RaQualia Pharma Industry-Academia Collaborative Research Center, Nagoya University Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Yukiko Moriguchi
- RaQualia Pharma Inc., RaQualia Pharma Industry-Academia Collaborative Research Center, Nagoya University Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Yusuke Ohno
- RaQualia Pharma Inc., RaQualia Pharma Industry-Academia Collaborative Research Center, Nagoya University Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Mio Yamasawa
- RaQualia Pharma Inc., RaQualia Pharma Industry-Academia Collaborative Research Center, Nagoya University Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Hirohide Noguchi
- RaQualia Pharma Inc., RaQualia Pharma Industry-Academia Collaborative Research Center, Nagoya University Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Akiyoshi Fujiuchi
- RaQualia Pharma Inc., RaQualia Pharma Industry-Academia Collaborative Research Center, Nagoya University Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Kazunobu Konishi
- Pharmaceuticals Research Centre, Asahi Kasei Pharma Corporation, 632-1, Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| |
Collapse
|
2
|
Merlin E, Salio C, Ferrini F. Painful Diabetic Neuropathy: Sex-Specific Mechanisms and Differences from Animal Models to Clinical Outcomes. Cells 2024; 13:2024. [PMID: 39682771 PMCID: PMC11640556 DOI: 10.3390/cells13232024] [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: 11/11/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Diabetes is a chronic and progressive disease associated with high blood glucose levels. Several co-morbidities arise from diabetes, the most common and severe one is diabetic neuropathy whose symptoms also include pain hypersensitivity. Currently, there are no effective therapies to counteract painful diabetic neuropathy or slow down the progression of the disease, and the underlying mechanisms are yet to be fully understood. Emerging data in recent decades have provided compelling evidence that the molecular and cellular mechanisms underlying chronic pain are different across the sexes. Interestingly, relevant differences have also been observed in the course and clinical presentation of painful diabetic neuropathy in humans. Here, we reviewed the current state of the art on sex differences in diabetic neuropathy, from animal models to clinical data. Comparing the output of both preclinical and clinical studies is necessary for properly orienting future choices in pain research, refining animal models, and interpreting clinical data. The identification of sex-specific mechanisms may help to develop more targeted therapies to counteract pain symptoms in diabetes.
Collapse
Affiliation(s)
- Emma Merlin
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095 Grugliasco, TO, Italy; (E.M.); (C.S.)
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095 Grugliasco, TO, Italy; (E.M.); (C.S.)
| | - Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095 Grugliasco, TO, Italy; (E.M.); (C.S.)
- Department of Psychiatry and Neurosciences, Université Laval, Québec, QC G1K 7P4, Canada
| |
Collapse
|
3
|
Soliman N, Kersebaum D, Lawn T, Sachau J, Sendel M, Vollert J. Improving neuropathic pain treatment - by rigorous stratification from bench to bedside. J Neurochem 2024; 168:3699-3714. [PMID: 36852505 DOI: 10.1111/jnc.15798] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/10/2023] [Accepted: 02/22/2023] [Indexed: 03/01/2023]
Abstract
Chronic pain is a constantly recurring and persistent illness, presenting a formidable healthcare challenge for patients and physicians alike. Current first-line analgesics offer only low-modest efficacy when averaged across populations, further contributing to this debilitating disease burden. Moreover, many recent trials for novel analgesics have not met primary efficacy endpoints, which is particularly striking considering the pharmacological advances have provided a range of highly relevant new drug targets. Heterogeneity within chronic pain cohorts is increasingly understood to play a critical role in these failures of treatment and drug discovery, with some patients deriving substantial benefits from a given intervention while it has little-to-no effect on others. As such, current treatment failures may not result from a true lack of efficacy, but rather a failure to target individuals whose pain is driven by mechanisms which it therapeutically modulates. This necessitates a move towards phenotypical stratification of patients to delineate responders and non-responders in a mechanistically driven manner. In this article, we outline a bench-to-bedside roadmap for this transition to mechanistically informed personalised pain medicine. We emphasise how the successful identification of novel analgesics is dependent on rigorous experimental design as well as the validity of models and translatability of outcome measures between the animal model and patients. Subsequently, we discuss general and specific aspects of human trial design to address heterogeneity in patient populations to increase the chance of identifying effective analgesics. Finally, we show how stratification approaches can be brought into clinical routine to the benefit of patients.
Collapse
Affiliation(s)
- Nadia Soliman
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Dilara Kersebaum
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Timothy Lawn
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Juliane Sachau
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Manon Sendel
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Jan Vollert
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, UK
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Germany
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
- Neurophysiology, Mannheim Center of Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
4
|
Bäckryd E, Themistocleous A, Larsson A, Gordh T, Rice ASC, Tesfaye S, Bennett DL, Gerdle B. Eleven neurology-related proteins measured in serum are positively correlated to the severity of diabetic neuropathy. Sci Rep 2024; 14:17068. [PMID: 39048581 PMCID: PMC11269577 DOI: 10.1038/s41598-024-66471-6] [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: 11/07/2023] [Accepted: 07/01/2024] [Indexed: 07/27/2024] Open
Abstract
About 20% of patients with diabetes suffer from chronic pain with neuropathic characteristics. We investigated the multivariate associations between 92 neurology-related proteins measured in serum from 190 patients with painful and painless diabetic neuropathy. Participants were recruited from the Pain in Neuropathy Study, an observational cross-sectional multicentre study in which participants underwent deep phenotyping. In the exploration cohort, two groups were defined by hierarchical cluster analyses of protein data. The proportion of painless vs painful neuropathy did not differ between the two groups, but one group had a significantly higher grade of neuropathy as measured by the Toronto Clinical Scoring System (TCSS). This finding was replicated in the replication cohort. Analyzing both groups together, we found that a group of 11 inter-correlated proteins (TNFRSF12A, SCARB2, N2DL-2, SKR3, EFNA4, LAYN, CLM-1, CD38, UNC5C, GFR-alpha-1, and JAM-B) were positively associated with TCSS values. Notably, EFNA4 and UNC5C are known to be part of axon guidance pathways. To conclude, although cluster analysis of 92 neurology-related proteins did not distinguish painful from painless diabetic neuropathy, we identified 11 proteins which positively correlated to neuropathy severity and warrant further investigation as potential biomarkers.
Collapse
Affiliation(s)
- Emmanuel Bäckryd
- Pain and Rehabilitation Center, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.
| | | | - Anders Larsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Torsten Gordh
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Andrew S C Rice
- Pain Research, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Solomon Tesfaye
- Diabetes Research Unit, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Björn Gerdle
- Pain and Rehabilitation Center, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
5
|
Vieira WF, Coelho DRA, Litwiler ST, McEachern KM, Clancy JA, Morales-Quezada L, Cassano P. Neuropathic pain, mood, and stress-related disorders: A literature review of comorbidity and co-pathogenesis. Neurosci Biobehav Rev 2024; 161:105673. [PMID: 38614452 DOI: 10.1016/j.neubiorev.2024.105673] [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: 01/30/2024] [Revised: 04/02/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Neuropathic pain can be caused by multiple factors, and its prevalence can reach 10% of the global population. It is becoming increasingly evident that limited or short-lasting response to treatments for neuropathic pain is associated with psychological factors, which include psychiatric comorbidities known to affect quality of life. It is estimated that 60% of patients with neuropathic pain also experience depression, anxiety, and stress symptoms. Altered mood, including stress, can be a consequence of several painful conditions but can also favor pain chronicization when preexisting. Despite the apparent tight connection between clinical pain and mood/stress disorders, the exact physiological mechanisms remain unclear. This review aims to provide an overview of state-of-the-art research on the mechanisms of pain related to the pathophysiology of depression, anxiety, and stress disorders.
Collapse
Affiliation(s)
- Willians Fernando Vieira
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA; Department of Psychiatry, Harvard Medical School (HMS), Boston, USA; Department of Anatomy, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), São Paulo, Brazil.
| | - David Richer Araujo Coelho
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA; Department of Psychiatry, Harvard Medical School (HMS), Boston, USA; Harvard T. H. Chan School of Public Health (HSPH), Boston, USA
| | - Scott Thomas Litwiler
- Center for Computational and Integrative Biology (CCIB), Massachusetts General Hospital (MGH), Boston, USA
| | - Kayla Marie McEachern
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA
| | - Julie A Clancy
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA
| | - Leon Morales-Quezada
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, USA
| | - Paolo Cassano
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA; Department of Psychiatry, Harvard Medical School (HMS), Boston, USA
| |
Collapse
|
6
|
Alexandre C, Miracca G, Holanda VD, Sharma A, Kourbanova K, Ferreira A, Bicca MA, Zeng X, Nassar VA, Lee S, Kaur S, Sarma SV, Sacré P, Scammell TE, Woolf CJ, Latremoliere A. Nociceptor spontaneous activity is responsible for fragmenting non-rapid eye movement sleep in mouse models of neuropathic pain. Sci Transl Med 2024; 16:eadg3036. [PMID: 38630850 PMCID: PMC11106840 DOI: 10.1126/scitranslmed.adg3036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/27/2024] [Indexed: 04/19/2024]
Abstract
Spontaneous pain, a major complaint of patients with neuropathic pain, has eluded study because there is no reliable marker in either preclinical models or clinical studies. Here, we performed a comprehensive electroencephalogram/electromyogram analysis of sleep in several mouse models of chronic pain: neuropathic (spared nerve injury and chronic constriction injury), inflammatory (Freund's complete adjuvant and carrageenan, plantar incision) and chemical pain (capsaicin). We find that peripheral axonal injury drives fragmentation of sleep by increasing brief arousals from non-rapid eye movement sleep (NREMS) without changing total sleep amount. In contrast to neuropathic pain, inflammatory or chemical pain did not increase brief arousals. NREMS fragmentation was reduced by the analgesics gabapentin and carbamazepine, and it resolved when pain sensitivity returned to normal in a transient neuropathic pain model (sciatic nerve crush). Genetic silencing of peripheral sensory neurons or ablation of CGRP+ neurons in the parabrachial nucleus prevented sleep fragmentation, whereas pharmacological blockade of skin sensory fibers was ineffective, indicating that the neural activity driving the arousals originates ectopically in primary nociceptor neurons and is relayed through the lateral parabrachial nucleus. These findings identify NREMS fragmentation by brief arousals as an effective proxy to measure spontaneous neuropathic pain in mice.
Collapse
Affiliation(s)
- Chloe Alexandre
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Giulia Miracca
- Department of Neurology, Beth israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- FM Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Victor Duarte Holanda
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ashley Sharma
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Kamila Kourbanova
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ashley Ferreira
- Department of Neurology, Beth israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- FM Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Maíra A. Bicca
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Xiangsunze Zeng
- FM Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Victoria A. Nassar
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Seungkyu Lee
- FM Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Satvinder Kaur
- Department of Neurology, Beth israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Sridevi V. Sarma
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Pierre Sacré
- Department of Electrical Engineering and Computer Science, School of Engineering, University of Liège, Liège, Belgium
| | - Thomas E. Scammell
- Department of Neurology, Beth israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Clifford J. Woolf
- FM Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Alban Latremoliere
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
7
|
Sant'Anna MB, Kimura LF, Vieira WF, Zambelli VO, Novaes LS, Hösch NG, Picolo G. Environmental factors and their impact on chronic pain development and maintenance. Phys Life Rev 2024; 48:176-197. [PMID: 38320380 DOI: 10.1016/j.plrev.2024.01.007] [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: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/08/2024]
Abstract
It is more than recognized and accepted that the environment affects the physiological responses of all living things, from bacteria to superior vertebrates, constituting an important factor in the evolution of all species. Environmental influences range from natural processes such as sunlight, seasons of the year, and rest to complex processes like stress and other mood disorders, infections, and air pollution, being all of them influenced by how each creature deals with them. In this chapter, it will be discussed how some of the environmental elements affect directly or indirectly neuropathic pain, a type of chronic pain caused by a lesion or disease of the somatosensory nervous system. For that, it was considered the edge of knowledge in translational research, thus including data from human and experimental animals as well as the applicability of such findings.
Collapse
Affiliation(s)
| | - Louise Faggionato Kimura
- Laboratory of Pain and Signaling, Butantan Institute, São Paulo, Brazil; Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, USA
| | - Willians Fernando Vieira
- Laboratory of Functional Neuroanatomy of Pain, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | | | - Leonardo Santana Novaes
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Gisele Picolo
- Laboratory of Pain and Signaling, Butantan Institute, São Paulo, Brazil.
| |
Collapse
|
8
|
Karimi SA, Zahra FT, Martin LJ. IUPHAR review: Navigating the role of preclinical models in pain research. Pharmacol Res 2024; 200:107073. [PMID: 38232910 DOI: 10.1016/j.phrs.2024.107073] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/19/2024]
Abstract
Chronic pain is a complex and challenging medical condition that affects millions of people worldwide. Understanding the underlying mechanisms of chronic pain is a key goal of preclinical pain research so that more effective treatment strategies can be developed. In this review, we explore nociception, pain, and the multifaceted factors that lead to chronic pain by focusing on preclinical models. We provide a detailed look into inflammatory and neuropathic pain models and discuss the most used animal models for studying the mechanisms behind these conditions. Additionally, we emphasize the vital role of these preclinical models in developing new pain-relief drugs, focusing on biologics and the therapeutic potential of NMDA and cannabinoid receptor antagonists. We also discuss the challenges of TRPV1 modulation for pain treatment, the clinical failures of neurokinin (NK)- 1 receptor antagonists, and the partial success story of Ziconotide to provide valuable lessons for preclinical pain models. Finally, we highlight the overall success and limitations of current treatments for chronic pain while providing critical insights into the development of more effective therapies to alleviate the burden of chronic pain.
Collapse
Affiliation(s)
- Seyed Asaad Karimi
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| | - Fatama Tuz Zahra
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Loren J Martin
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada.
| |
Collapse
|
9
|
Soliman N, Denk F. Practical approaches to improving translatability and reproducibility in preclinical pain research. Brain Behav Immun 2024; 115:38-42. [PMID: 37793487 DOI: 10.1016/j.bbi.2023.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/21/2023] [Accepted: 09/30/2023] [Indexed: 10/06/2023] Open
Abstract
Pain research continues to face the challenge of poor translatability of pre-clinical studies. In this short primer, we are summarizing the possible causes, with an emphasis on practical and constructive solutions. In particular, we stress the importance of increased heterogeneity in animal studies; formal or informal pre-registration to combat publication bias; and increased statistical training in order to help pre-clinical scientists appreciate the usefulness of available experimental design and reporting guidelines.
Collapse
Affiliation(s)
- Nadia Soliman
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Franziska Denk
- Wolfson Centre for Age-related Diseases, King's College London, Guy's Campus, London, SE1 1UL, United Kingdom.
| |
Collapse
|
10
|
Shepherd AJ, Rice AS, Smith MT. Angiotensin II type 2 receptor signalling as a pain target: Bench, bedside and back-translation. Curr Opin Pharmacol 2023; 73:102415. [PMID: 38041933 PMCID: PMC11789660 DOI: 10.1016/j.coph.2023.102415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 12/04/2023]
Abstract
Translating promising preclinical pain relief data for novel molecules from drug discovery to positive clinical trial outcomes is challenging. The angiotensin II type 2 (AT2) receptor is a clinically-validated target based upon positive proof-of-concept clinical trial data in patients with post-herpetic neuralgia. This trial was conducted because AT2 receptor antagonists evoked pain relief in rodent models of neuropathic pain. EMA401 was selected as the drug candidate based upon its suitable preclinical toxicity and safety profile and good pharmacokinetics. Herein, we provide an overview of the discovery, preclinical and clinical development of EMA401, for the alleviation of peripheral neuropathic pain.
Collapse
Affiliation(s)
- Andrew J Shepherd
- The MD Anderson Pain Research Consortium and the Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew Sc Rice
- Pain Research, Department of Surgery & Cancer, Imperial College London, London, UK
| | - Maree T Smith
- School of Biomedical Sciences, The University of Queensland, St Lucia Campus, Brisbane, Queensland, Australia.
| |
Collapse
|
11
|
Smith PA. Neuropathic pain; what we know and what we should do about it. FRONTIERS IN PAIN RESEARCH 2023; 4:1220034. [PMID: 37810432 PMCID: PMC10559888 DOI: 10.3389/fpain.2023.1220034] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Neuropathic pain can result from injury to, or disease of the nervous system. It is notoriously difficult to treat. Peripheral nerve injury promotes Schwann cell activation and invasion of immunocompetent cells into the site of injury, spinal cord and higher sensory structures such as thalamus and cingulate and sensory cortices. Various cytokines, chemokines, growth factors, monoamines and neuropeptides effect two-way signalling between neurons, glia and immune cells. This promotes sustained hyperexcitability and spontaneous activity in primary afferents that is crucial for onset and persistence of pain as well as misprocessing of sensory information in the spinal cord and supraspinal structures. Much of the current understanding of pain aetiology and identification of drug targets derives from studies of the consequences of peripheral nerve injury in rodent models. Although a vast amount of information has been forthcoming, the translation of this information into the clinical arena has been minimal. Few, if any, major therapeutic approaches have appeared since the mid 1990's. This may reflect failure to recognise differences in pain processing in males vs. females, differences in cellular responses to different types of injury and differences in pain processing in humans vs. animals. Basic science and clinical approaches which seek to bridge this knowledge gap include better assessment of pain in animal models, use of pain models which better emulate human disease, and stratification of human pain phenotypes according to quantitative assessment of signs and symptoms of disease. This can lead to more personalized and effective treatments for individual patients. Significance statement: There is an urgent need to find new treatments for neuropathic pain. Although classical animal models have revealed essential features of pain aetiology such as peripheral and central sensitization and some of the molecular and cellular mechanisms involved, they do not adequately model the multiplicity of disease states or injuries that may bring forth neuropathic pain in the clinic. This review seeks to integrate information from the multiplicity of disciplines that seek to understand neuropathic pain; including immunology, cell biology, electrophysiology and biophysics, anatomy, cell biology, neurology, molecular biology, pharmacology and behavioral science. Beyond this, it underlines ongoing refinements in basic science and clinical practice that will engender improved approaches to pain management.
Collapse
Affiliation(s)
- Peter A. Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
12
|
Zhang XY, Diaz-delCastillo M, Kong L, Daniels N, MacIntosh-Smith W, Abdallah A, Domanski D, Sofrenovic D, Yeung TP(S, Valiente D, Vollert J, Sena E, Rice AS, Soliman N. A systematic review and meta-analysis of thigmotactic behaviour in the open field test in rodent models associated with persistent pain. PLoS One 2023; 18:e0290382. [PMID: 37682863 PMCID: PMC10490990 DOI: 10.1371/journal.pone.0290382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/07/2023] [Indexed: 09/10/2023] Open
Abstract
Thigmotaxis is an innate predator avoidance behaviour of rodents. To gain insight into how injury and disease models, and analgesic drug treatments affect thigmotaxis, we performed a systematic review and meta-analysis of studies that assessed thigmotaxis in the open field test. Systematic searches were conducted of 3 databases in October 2020, March and August 2022. Study design characteristics and experimental data were extracted and analysed using a random-effects meta-analysis. We also assessed the correlation between thigmotaxis and stimulus-evoked limb withdrawal. This review included the meta-analysis of 165 studies We report thigmotaxis was increased in injury and disease models associated with persistent pain and this increase was attenuated by analgesic drug treatments in both rat and mouse experiments. Its usefulness, however, may be limited in certain injury and disease models because our analysis suggested that thigmotaxis may be associated with the locomotor function. We also conducted subgroup analyses and meta-regression, but our findings on sources of heterogeneity are inconclusive because analyses were limited by insufficient available data. It was difficult to assess internal validity because reporting of methodological quality measures was poor, therefore, the studies have an unclear risk of bias. The correlation between time in the centre (type of a thigmotactic metric) and types of stimulus-evoked limb withdrawal was inconsistent. Therefore, stimulus-evoked and ethologically relevant behavioural paradigms should be viewed as two separate entities as they are conceptually and methodologically different from each other.
Collapse
Affiliation(s)
- Xue Ying Zhang
- Pain Research, Department of Surgery and Cancer, Imperial College London, United Kingdom
| | | | - Lingsi Kong
- Department of Women and Children’s Health, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
| | - Natasha Daniels
- Bart’s Health NHS Trust Whipps Cross Hospital, London, United Kingdom
| | - William MacIntosh-Smith
- School of Medicine, Medical Science and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Aya Abdallah
- School of Medicine, Medical Science and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Dominik Domanski
- School of Medicine, Medical Science and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Denis Sofrenovic
- Faculty of Medicine, Imperial College London, London, United Kingdom
| | | | - Diego Valiente
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Jan Vollert
- Pain Research, Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Emily Sena
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew S. Rice
- Pain Research, Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Nadia Soliman
- Pain Research, Department of Surgery and Cancer, Imperial College London, United Kingdom
| |
Collapse
|
13
|
Mansfield M, Thacker M. Integrating jigsaw puzzle thinking into practice: the assessment of cervical spine radiculopathy. Curr Opin Support Palliat Care 2023; 17:135-141. [PMID: 37389587 DOI: 10.1097/spc.0000000000000656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
PURPOSE OF REVIEW Cervical spine radiculopathy (CSR) presents a complex socioeconomic problem for patients, clinicians, families, employers and healthcare systems. Due to the heterogeneity of clinical presentation and underlying mechanisms, clinical assessment can be challenging. This review will examine the literature on the underlying pathophysiology and studies investigating the holistic assessment strategies for this disabling condition. The authors will focus particular attention on the psychological factors associated with CSR and the physical and imaging strategies to establish a diagnosis. RECENT FINDINGS Contemporary CSR assessment should identify the underlying pathomechanisms and how this may impact the somatosensory nervous system integrity and function. No physical assessment test in isolation will establish CSR diagnosis; therefore, clinicians should utilise a cluster of tests and recognise the potential limitations as part of a clinical reasoning framework. The assessment of the somatosensory nervous system can provide insights into particular subgroups of CSR presentation, which may provide interesting opportunities to continue to enhance individualised assessment and management strategies for CSR. The interplay between psychological factors can influence the diagnosis and recovery times for a person with CSR, and clinicians should continue to explore how these factors may influence a person's prognosis. The authors will discuss the opportunities for future research and limitations of contemporary approaches to assessment, underpinned by evidence, and how this supports a clinical assessment to establish CSR diagnosis. SUMMARY Research should continue to investigate how clinicians assess the interplay between physical and psychological factors to inform the establishment of CSR. Specifically, there is a need to investigate the validity and reliability of combining somatosensory, motor and imaging assessment findings to reach a diagnosis and inform onward management plans.
Collapse
Affiliation(s)
- Michael Mansfield
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston
- Centre of Precision Rehabilitation for Spinal Pain, University of Birmingham, Birmingham, UK
| | - Mick Thacker
- School of Physiotherapy, Royal College of Surgeons Ireland, Dublin 2, Ireland
| |
Collapse
|
14
|
Edwards RR, Schreiber KL, Dworkin RH, Turk DC, Baron R, Freeman R, Jensen TS, Latremoliere A, Markman JD, Rice ASC, Rowbotham M, Staud R, Tate S, Woolf CJ, Andrews NA, Carr DB, Colloca L, Cosma-Roman D, Cowan P, Diatchenko L, Farrar J, Gewandter JS, Gilron I, Kerns RD, Marchand S, Niebler G, Patel KV, Simon LS, Tockarshewsky T, Vanhove GF, Vardeh D, Walco GA, Wasan AD, Wesselmann U. Optimizing and Accelerating the Development of Precision Pain Treatments for Chronic Pain: IMMPACT Review and Recommendations. THE JOURNAL OF PAIN 2023; 24:204-225. [PMID: 36198371 PMCID: PMC10868532 DOI: 10.1016/j.jpain.2022.08.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/01/2022] [Accepted: 08/17/2022] [Indexed: 11/06/2022]
Abstract
Large variability in the individual response to even the most-efficacious pain treatments is observed clinically, which has led to calls for a more personalized, tailored approach to treating patients with pain (ie, "precision pain medicine"). Precision pain medicine, currently an aspirational goal, would consist of empirically based algorithms that determine the optimal treatments, or treatment combinations, for specific patients (ie, targeting the right treatment, in the right dose, to the right patient, at the right time). Answering this question of "what works for whom" will certainly improve the clinical care of patients with pain. It may also support the success of novel drug development in pain, making it easier to identify novel treatments that work for certain patients and more accurately identify the magnitude of the treatment effect for those subgroups. Significant preliminary work has been done in this area, and analgesic trials are beginning to utilize precision pain medicine approaches such as stratified allocation on the basis of prespecified patient phenotypes using assessment methodologies such as quantitative sensory testing. Current major challenges within the field include: 1) identifying optimal measurement approaches to assessing patient characteristics that are most robustly and consistently predictive of inter-patient variation in specific analgesic treatment outcomes, 2) designing clinical trials that can identify treatment-by-phenotype interactions, and 3) selecting the most promising therapeutics to be tested in this way. This review surveys the current state of precision pain medicine, with a focus on drug treatments (which have been most-studied in a precision pain medicine context). It further presents a set of evidence-based recommendations for accelerating the application of precision pain methods in chronic pain research. PERSPECTIVE: Given the considerable variability in treatment outcomes for chronic pain, progress in precision pain treatment is critical for the field. An array of phenotypes and mechanisms contribute to chronic pain; this review summarizes current knowledge regarding which treatments are most effective for patients with specific biopsychosocial characteristics.
Collapse
Affiliation(s)
| | | | | | - Dennis C Turk
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
| | - Ralf Baron
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Arnold-Heller-Straße 3, House D, 24105 Kiel, Germany
| | - Roy Freeman
- Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | | | | | | | - Nick A Andrews
- Salk Institute for Biological Studies, San Diego, California
| | | | | | | | - Penney Cowan
- American Chronic Pain Association, Rocklin, California
| | - Luda Diatchenko
- Department of Anesthesia and Faculty of Dentistry, McGill University, Montreal, California
| | - John Farrar
- University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Robert D Kerns
- Yale University, Departments of Psychiatry, Neurology, and Psychology, New Haven, Connecticut
| | | | | | - Kushang V Patel
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
| | | | | | | | | | - Gary A Walco
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
| | - Ajay D Wasan
- University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ursula Wesselmann
- Department of Anesthesiology/Division of Pain Medicine, Neurology and Psychology, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
15
|
Natale CA, Christie MJ, Aubrey KR. Spinal glycinergic currents are reduced in a rat model of neuropathic pain following partial nerve ligation but not chronic constriction injury. J Neurophysiol 2023; 129:333-341. [PMID: 36541621 DOI: 10.1152/jn.00451.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Animal models have consistently indicated that central sensitization and the development of chronic neuropathic pain are linked to changes to inhibitory signaling in the dorsal horn of the spinal cord. However, replication of data investigating the cellular mechanisms that underlie these changes remains a challenge and there is still a lack of understanding about what aspects of spinal inhibitory transmission most strongly contribute to the disease. Here, we compared the effect of two different sciatic nerve injuries commonly used to generate rodent models of neuropathic pain on spinal glycinergic signaling. Using whole cell patch-clamp electrophysiology in spinal slices, we recorded from neurons in the lamina II of the dorsal horn and evoked inhibitory postsynaptic currents with a stimulator in lamina III, where glycinergic cell bodies are concentrated. We found that glycine inputs onto radial neurons were reduced following partial nerve ligation (PNL) of the sciatic nerve, consistent with a previous report. However, this finding was not replicated in animals that underwent chronic constriction injury (CCI) to the same nerve region. To limit the between-experiment variability, we kept the rat species, sex, and age consistent and had a single investigator carry out the surgeries. These data show that PNL and CCI cause divergent spinal signaling outcomes in the cord and add to the body of evidence suggesting that treatments for neuropathic pain should be triaged according to nerve injury or cellular dysfunction rather than the symptoms of the disease.NEW & NOTEWORTHY Neuropathic pain models are used in preclinical research to investigate the mechanisms underlying allodynia, a common symptom of neuropathic pain, and to test, develop, and validate therapies for persistent pain. We demonstrate that a glycinergic dysfunction is consistently associated with partial nerve ligation but not the chronic constriction injury model. This suggests that the cellular effects produced by each injury are distinct and that data from different neuropathic pain models should be considered separately.
Collapse
Affiliation(s)
- Claudia A Natale
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Macdonald J Christie
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Karin R Aubrey
- Pain Management Research, Kolling Institute, Royal North Shore Hospital, St Leonards, New South Wales, Australia.,Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
16
|
Zhang XY, Barakat A, Diaz-delCastillo M, Vollert J, Sena ES, Heegaard AM, Rice AS, Soliman N. Systematic review and meta-analysis of studies in which burrowing behaviour was assessed in rodent models of disease-associated persistent pain. Pain 2022; 163:2076-2102. [PMID: 35353780 PMCID: PMC9578533 DOI: 10.1097/j.pain.0000000000002632] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 12/09/2022]
Abstract
ABSTRACT Burrowing behaviour is used to assess pain-associated behaviour in laboratory rodents. To gain insight into how models of disease-associated persistent pain and analgesics affect burrowing behaviour, we performed a systematic review and meta-analysis of studies that assessed burrowing behaviour. A systematic search in March 2020 and update in September 2020 was conducted in 4 databases. Study design characteristics and experimental data were extracted, followed by a random-effects meta-analysis. We explored the association between burrowing and monofilament-induced limb withdrawal. Dose response relationship was investigated for some analgesics. Forty-five studies were included in the meta-analysis, in which 16 model types and 14 drug classes were used. Most experiments used rat (79%) and male (72%) animals. Somatic inflammation and trauma-induced neuropathy models were associated with reduced burrowing behaviour. Analgesics (nonsteroidal anti-inflammatory drug and gabapentinoids) attenuated burrowing deficits in these models. Reporting of measures to reduce risk of bias was unclear except for randomisation which was high. There was not a correlation ( R2 = 0.1421) between burrowing and monofilament-induced limb withdrawal. Opioids, gabapentin, and naproxen showed reduced burrowing behaviour at high doses, whereas ibuprofen and celecoxib showed opposite trend. The findings indicate that burrowing could be used to assess pain-associated behaviour. We support the use of a portfolio of composite measures including spontaneous and stimulus-evoked tests. The information collected here could help in designing experiments involving burrowing assessment in models of disease-associated pain.
Collapse
Affiliation(s)
- Xue Ying Zhang
- Pain Research, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Ahmed Barakat
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Marta Diaz-delCastillo
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jan Vollert
- Pain Research, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital of Schleswig-Holstein, Campus Kiel, Germany
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Germany
- Neurophysiology, Mannheim Centre of Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Germany
| | - Emily S. Sena
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Anne-Marie Heegaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrew S.C. Rice
- Pain Research, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Nadia Soliman
- Pain Research, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
17
|
Matesanz-García L, Schmid AB, Cáceres-Pajuelo JE, Cuenca-Martínez F, Arribas-Romano A, González-Zamorano Y, Goicoechea-García C, Fernández-Carnero J. Effect of Physiotherapeutic Interventions on Biomarkers of Neuropathic Pain: A Systematic Review of Preclinical Literature. THE JOURNAL OF PAIN 2022; 23:1833-1855. [PMID: 35768044 PMCID: PMC7613788 DOI: 10.1016/j.jpain.2022.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 02/02/2023]
Abstract
The purpose of this systematic review was to evaluate the effects of physiotherapeutic interventions on biomarkers of neuropathic pain in preclinical models of peripheral neuropathic pain (PNP). The search was performed in Pubmed, Web of Science, EMBASE, Cochrane, Cinhal, Psycinfo, Scopus, Medline, and Science Direct. Studies evaluating any type of physiotherapy intervention for PNP (systemic or traumatic) were included. Eighty-one articles were included in this review. The most common PNP model was chronic constriction injury, and the most frequently studied biomarkers were related to neuro-immune processes. Exercise therapy and Electro-acupuncture were the 2 most frequently studied physiotherapy interventions while acupuncture and joint mobilization were less frequently examined. Most physiotherapeutic interventions modulated the expression of biomarkers related to neuropathic pain. Whereas the results seem promising; they have to be considered with caution due to the high risk of bias of included studies and high heterogeneity of the type and anatomical localization of biomarkers reported. The review protocol is registered on PROSPERO (CRD42019142878). PERSPECTIVE: This article presents the current evidence about physiotherapeutic interventions on biomarkers of neuropathic pain in preclinical models of peripheral neuropathic pain. Existing findings are reviewed, and relevant data are provided on the effectiveness of each physiotherapeutic modality, as well as its certainty of evidence and clinical applicability.
Collapse
Affiliation(s)
- Luis Matesanz-García
- Escuela Internacional de Doctorado, Department of Physical Therapy, Occupational Therapy, Rehabilitation and Physical Medicine, Universidad Rey Juan Carlos, Alcorcón, Spain; Departamento de Fisioterapia, Centro Superior de Estudios Universitarios La Salle, Universidad Autónoma de Madrid, Madrid, Spain
| | - Annina B Schmid
- Nuffield Department for Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | | | - Ferran Cuenca-Martínez
- Exercise Intervention for Health Research Group (EXINH-RG), Department of Physiotherapy, University of Valencia, Valencia, Spain.
| | - Alberto Arribas-Romano
- Escuela Internacional de Doctorado, Department of Physical Therapy, Occupational Therapy, Rehabilitation and Physical Medicine, Universidad Rey Juan Carlos, Alcorcón, Spain; Department of Physical Therapy, Occupational Therapy, Rehabilitation and Physical Medicine, Rey Juan Carlos University, Madrid, Spain
| | - Yeray González-Zamorano
- Escuela Internacional de Doctorado, Department of Physical Therapy, Occupational Therapy, Rehabilitation and Physical Medicine, Universidad Rey Juan Carlos, Alcorcón, Spain; Grupo de Investigación de Neurorrehabilitación del Daño Cerebral y los Trastornos del Movimiento (GINDAT), Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | | | - Josué Fernández-Carnero
- Department of Physical Therapy, Occupational Therapy, Rehabilitation and Physical Medicine, Rey Juan Carlos University, Madrid, Spain; Grupo de Investigación de Neurorrehabilitación del Daño Cerebral y los Trastornos del Movimiento (GINDAT), Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain; Motion in Brains Research Group, Institute of Neuroscience and Sciences of the Movement (INCIMOV), Centro Superior de Estudios Universitarios La Salle, Universidad Autónoma de Madrid, Madrid, Spain; Grupo Multidisciplinar de Investigación y Tratamiento del Dolor, Grupo de Excelencia Investigadora URJC-Banco de Santander, Madrid, Spain; La Paz Hospital Institute for Health Research, IdiPAZ, Madrid, Spain
| |
Collapse
|
18
|
Coexistence of chronic hyperalgesia and multilevel neuroinflammatory responses after experimental SCI: a systematic approach to profiling neuropathic pain. J Neuroinflammation 2022; 19:264. [PMID: 36309729 PMCID: PMC9617391 DOI: 10.1186/s12974-022-02628-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/20/2022] [Indexed: 11/11/2022] Open
Abstract
Background People with spinal cord injury (SCI) frequently develop neuropathic pain (NP) that worsens disability and diminishes rehabilitation efficacy. Chronic NP is presently incurable due to poor understanding of underlying mechanisms. We hypothesized that multilocus neuroinflammation (NIF) might be a driver of SCI NP, and tested it by investigating whether NP coexisted with central NIF, neurotransmission (NTM), neuromodulation (NML) and neuroplasticity (NPL) changes post-SCI. Methods Female Sprague–Dawley rats (230–250 g) with T10 compression or laminectomy were evaluated for physical conditions, coordinated hindlimb functions, neurological reflexes, and mechanical/thermal sensitivity thresholds at 1 day post-injury (p.i.) and weekly thereafter. Eight weeks p.i., central nervous system tissues were histochemically and immunohistochemically characterized for parameters/markers of histopathology and NIF/NTM/NML/NPL. Also analyzed was the correlative relationship between levels of selected biomarkers and thermosensitivity thresholds via statistical linear regression. Results SCI impaired sensorimotor functions, altered reflexes, and produced spontaneous pain signs and hypersensitivity to evoked nociceptive, mechanical, and thermal inputs. Only injured spinal cords exhibited neural lesion, microglia/astrocyte activation, and abnormal expression of proinflammatory cytokines, as well as NIF/NTM/NML/NPL markers. Brains of SCI animals displayed similar pathophysiological signs in the gracile and parabrachial nuclei (GrN and PBN: sensory relay), raphe magnus nucleus and periaqueduct gray (RMN and PAG: pain modulation), basolateral amygdala (BLA: emotional-affective dimension of pain), and hippocampus (HPC: memory/mood/neurogenesis). SCI augmented sensory NTM/NPL (GrN and PBN); increased GAD67 (PAG) level; reduced serotonin (RMN) and fear-off neuronal NTR2 (BLA) expressions; and perturbed neurogenesis (HPC). Conclusion T10 compression caused chronic hyperalgesia that coexisted with NIF/NTM/NML/NPL responses at multilevel neuroaxis centers. The data have provided multidimensional biomarkers as new mechanistic leads to profile SCI NP for therapeutic/therapy development. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02628-2.
Collapse
|
19
|
Gewandter JS, Sohn MB, De Guzman R, Frazer ME, Chiodo V, Sharma S, Geha P, Markman JD. Predicting Treatment Response with Sensory Phenotyping in Post-Traumatic Neuropathic Pain. PAIN MEDICINE (MALDEN, MASS.) 2022; 23:1726-1732. [PMID: 35312012 PMCID: PMC9527609 DOI: 10.1093/pm/pnac045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Currently available treatments for neuropathic pain are only modestly efficacious when assessed in randomized clinical trials and work for only some patients in the clinic. Induced-pain or gain-of-function phenotypes have been shown to predict response to analgesics (vs placebos) in patients with neuropathic pain. However, the predictive value of these phenotypes has never been studied in post-traumatic neuropathic pain. METHODS Mixed-effects models for repeated measures were used to evaluate the efficacy of pregabalin vs placebo in subgroups with induced-pain phenotypes (i.e., hyperalgesia or allodynia) in data from a recent, multinational randomized clinical trial (N = 539) that identified phenotypic subgroups through the use of a structured clinical exam. RESULTS The difference in mean pain score between the active and placebo groups (i.e., delta) after 15 weeks of treatment for the subgroup with hyperalgesia was -0.76 (P = 0.001), compared with 0.19 (P = 0.47) for the subgroup that did not have hyperalgesia. The treatment-by-phenotype interaction, which tests whether subgroups have statistically different treatment responses, was significant (P = 0.0067). The delta for the subgroup with allodynia was -0.31 (P = 0.22), compared with -0.30 (P = 0.22) for the subgroup that did not have allodynia (treatment-by-phenotype interaction P = 0.98). CONCLUSIONS These data suggest that hyperalgesia, but not allodynia, predicts response to pregabalin in patients with chronic post-traumatic neuropathic pain. This study extends the growing data supporting the utility of induced-pain phenotypes to predict response to analgesics in post-traumatic neuropathic pain. Sensory phenotyping in large, multisite trials through the use of a structured clinical exam has the potential to accelerate the development of new analgesics and improve the generalizability of clinical trial results.
Collapse
Affiliation(s)
- Jennifer S Gewandter
- Correspondence to: Jennifer S. Gewandter, PhD, MPH, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave, Box 604, Rochester, NY 14642, USA. Tel: 585-276-5661; Fax: 585-244-7271; E-mail:
| | - Michael B Sohn
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York
| | - Rachel De Guzman
- Department of Neurosurgery, University of Rochester, Rochester, New York
| | - Maria E Frazer
- Department of Neurosurgery, University of Rochester, Rochester, New York
| | - Valerie Chiodo
- Department of Neurosurgery, University of Rochester, Rochester, New York
| | - Sonia Sharma
- Department of Oral Diagnostic Sciences, University at Buffalo School of Dental Medicine, Buffalo, New York
| | - Paul Geha
- Department of Psychiatry, University of Rochester, Rochester, New York, USA
| | - John D Markman
- Department of Neurosurgery, University of Rochester, Rochester, New York
| |
Collapse
|
20
|
Hargreaves R, Akinsanya K, Ajit SK, Dhruv NT, Driscoll J, Farina P, Gavva N, Gill M, Houghton A, Iyengar S, Jones C, Kavelaars A, Kaykas A, Koroshetz WJ, Laeng P, Laird JM, Lo DC, Luthman J, Munro G, Oshinsky ML, Sittampalam GS, Woller SA, Tamiz AP. Preclinical target validation for non-addictive therapeutics development for pain. Expert Opin Ther Targets 2022; 26:811-822. [DOI: 10.1080/14728222.2022.2147063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
| | | | - Seena K. Ajit
- Department of Pharmacology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States
| | - Neel T. Dhruv
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - Jamie Driscoll
- National Institute of Mental Health, Bethesda, Maryland, United States
| | - Peter Farina
- Canaan Partners, Westport, Connecticut, United States
| | - Narender Gavva
- Drug Discovery Sciences, Takeda Pharmaceuticals, San Diego, California, United States
| | - Marie Gill
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | | | - Smriti Iyengar
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - Carrie Jones
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States
| | - Annemieke Kavelaars
- The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | | | - Walter J. Koroshetz
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - Pascal Laeng
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - Jennifer M. Laird
- Eli Lilly and Company, Windlesham, United Kingdom of Great Britain and Northern Ireland
| | - Donald C. Lo
- National Center for Advancing Translational Sciences, Bethesda, Maryland, United States
| | | | | | - Michael L. Oshinsky
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - G. Sitta Sittampalam
- National Center for Advancing Translational Sciences, Bethesda, Maryland, United States
| | - Sarah A. Woller
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - Amir P. Tamiz
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| |
Collapse
|
21
|
Ameenudeen S, Kashif M, Banerjee S, Srinivasan H, Pandurangan AK, Waseem M. Cellular and Molecular Machinery of Neuropathic Pain: an Emerging Insight. CURRENT PHARMACOLOGY REPORTS 2022; 8:227-235. [PMID: 35646513 PMCID: PMC9125010 DOI: 10.1007/s40495-022-00294-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/17/2022] [Indexed: 12/04/2022]
Abstract
Purpose of Review Neuropathic pain (NP) has been ubiquitously characterized by lesion and its linked somatosensory system either the central nervous system (CNS) or peripheral nervous system (PNS) This PNS episode is the most prevalent site of NP origin and is found to be associated with afferent nerve fibers carrying pain signals from injured/trauma site to the CNS including the brain. Several kinds of pharmacotherapeutic drugs shuch as analgesics, anti-convulsants, and anti-depressants are being employed for the its possible interventions. The NP has been a great interest to follow different pathophysiological mechanisms which are often considered to correlate with the metabolic pathways and its mediated disease. There is paucity of knowledge to make such mechanism via NP. Recent Finding Most notably, recent pandemic outbreak of COVID-19 has also been reported in chronic pain mediated diabetes, inflammatory disorders, and cancers. There is an increasing incidence of NP and its complex mechanism has now led to identify the possible investigations of responsible genes and proteins via bioinformatics tools. The analysis might be more instrumental as collecting the genes from pain genetic database, analyzing the variants through differential gene expression (DEG) and constructing the protein–protein interaction (PPI) networks and thereby determining their upregulating and downregulating pathways. Summary This review sheds a bright light towards several mechanisms at both cellular and molecular level, correlation of NP-mediated disease mechanism and possible cell surface biomarkers (receptors), and identified genes could be more promising for their pharmacological targets.
Collapse
Affiliation(s)
- Shabnam Ameenudeen
- School of Life Sciences, BS Abdur Rahman Crescent Institute of Science and Technology, GST Road, Vandalur, Chennai, 600048 Tamil Nadu India
| | - Mohd. Kashif
- School of Life Sciences, BS Abdur Rahman Crescent Institute of Science and Technology, GST Road, Vandalur, Chennai, 600048 Tamil Nadu India
| | - Subhamoy Banerjee
- Department of Basic Science and Humanities, Institute of Engineering and Management, Sector V, Salt Lake, Kolkata, 700091 India
| | - Hemalatha Srinivasan
- School of Life Sciences, BS Abdur Rahman Crescent Institute of Science and Technology, GST Road, Vandalur, Chennai, 600048 Tamil Nadu India
| | - Ashok Kumar Pandurangan
- School of Life Sciences, BS Abdur Rahman Crescent Institute of Science and Technology, GST Road, Vandalur, Chennai, 600048 Tamil Nadu India
| | - Mohammad Waseem
- School of Life Sciences, BS Abdur Rahman Crescent Institute of Science and Technology, GST Road, Vandalur, Chennai, 600048 Tamil Nadu India
| |
Collapse
|
22
|
Zhang XY, Vollert J, Sena ES, Rice AS, Soliman N. A protocol for the systematic review and meta-analysis of thigmotactic behaviour in the open field test in rodent models associated with persistent pain. BMJ OPEN SCIENCE 2022; 5:e100135. [PMID: 35047702 PMCID: PMC8647568 DOI: 10.1136/bmjos-2020-100135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/30/2020] [Accepted: 12/17/2020] [Indexed: 12/09/2022] Open
Abstract
Objective Thigmotaxis is an innate predator avoidance behaviour of rodents and is enhanced when animals are under stress. It is characterised by the preference of a rodent to seek shelter, rather than expose itself to the aversive open area. The behaviour has been proposed to be a measurable construct that can address the impact of pain on rodent behaviour. This systematic review will assess whether thigmotaxis can be influenced by experimental persistent pain and attenuated by pharmacological interventions in rodents. Search strategy We will conduct search on three electronic databases to identify studies in which thigmotaxis was used as an outcome measure contextualised to a rodent model associated with persistent pain. All studies published until the date of the search will be considered. Screening and annotation Two independent reviewers will screen studies based on the order of (1) titles and abstracts, and (2) full texts. Data management and reporting For meta-analysis, we will extract thigmotactic behavioural data and calculate effect sizes. Effect sizes will be combined using a random-effects model. We will assess heterogeneity and identify sources of heterogeneity. A risk-of-bias assessment will be conducted to evaluate study quality. Publication bias will be assessed using funnel plots, Egger’s regression and trim-and-fill analysis. We will also extract stimulus-evoked limb withdrawal data to assess its correlation with thigmotaxis in the same animals. The evidence obtained will provide a comprehensive understanding of the strengths and limitations of using thigmotactic outcome measure in animal pain research so that future experimental designs can be optimised. We will follow the Preferred Reporting Items for Systematic Reviews and Meta-Analyses reporting guidelines and disseminate the review findings through publication and conference presentation.
Collapse
Affiliation(s)
| | - Jan Vollert
- Musculoskeletal, Imperial College London, London, UK
| | - Emily S Sena
- Clinical Neurosciences, University of Edinburgh, Edinburgh, UK
| | | | - Nadia Soliman
- Musculoskeletal, Imperial College London, London, UK
| |
Collapse
|
23
|
Démosthènes A, Sion B, Giraudet F, Moisset X, Daulhac L, Eschalier A, Bégou M. In-Depth Characterization of Somatic and Orofacial Sensitive Dysfunctions and Interfering-Symptoms in a Relapsing-Remitting Experimental Autoimmune Encephalomyelitis Mouse Model. Front Neurol 2022; 12:789432. [PMID: 35111128 PMCID: PMC8801881 DOI: 10.3389/fneur.2021.789432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/16/2021] [Indexed: 11/23/2022] Open
Abstract
Among the many symptoms (motor, sensory, and cognitive) associated with multiple sclerosis (MS), chronic pain is a common disabling condition. In particular, neuropathic pain symptoms are very prevalent and debilitating, even in early stages of the disease. Unfortunately, chronic pain still lacks efficient therapeutic agents. Progress is needed (i) clinically by better characterizing pain symptoms in MS and understanding the underlying mechanisms, and (ii) preclinically by developing a more closely dedicated model to identify new therapeutic targets and evaluate new drugs. In this setting, new variants of experimental autoimmune encephalomyelitis (EAE) are currently developed in mice to exhibit less severe motor impairments, thereby avoiding confounding factors in assessing pain behaviors over the disease course. Among these, the optimized relapsing-remitting EAE (QuilA-EAE) mouse model, induced using myelin oligodendrocyte glycoprotein peptide fragment (35–55), pertussis toxin, and quillaja bark saponin, seems very promising. Our study sought (i) to better define sensitive dysfunctions and (ii) to extend behavioral characterization to interfering symptoms often associated with pain during MS, such as mood disturbances, fatigue, and cognitive impairment, in this optimized QuilA-EAE model. We made an in-depth characterization of this optimized QuilA-EAE model, describing for the first time somatic thermal hyperalgesia associated with mechanical and cold allodynia. Evaluation of orofacial pain sensitivity showed no mechanical or thermal allodynia. Detailed evaluation of motor behaviors highlighted slight defects in fine motor coordination in the QuilA-EAE mice but without impact on pain evaluation. Finally, no anxiety-related or cognitive impairment was observed during the peak of sensitive symptoms. Pharmacologically, as previously described, we found that pregabalin, a treatment commonly used in neuropathic pain patients, induced an analgesic effect on mechanical allodynia. In addition, we showed an anti-hyperalgesic thermal effect on this model. Our results demonstrate that this QuilA-EAE model is clearly of interest for studying pain symptom development and so could be used to identify and evaluate new therapeutic targets. The presence of interfering symptoms still needs to be further characterized.
Collapse
Affiliation(s)
- Amélie Démosthènes
- Université Clermont Auvergne, Inserm, Neuro-Dol, Faculté de Pharmacie, Faculté de Médecine, Institut Analgesia, BP38, Clermont-Ferrand, France
| | - Benoît Sion
- Université Clermont Auvergne, Inserm, Neuro-Dol, Faculté de Pharmacie, Faculté de Médecine, Institut Analgesia, BP38, Clermont-Ferrand, France
| | - Fabrice Giraudet
- Université Clermont Auvergne, Inserm, Neuro-Dol, Faculté de Pharmacie, Faculté de Médecine, Institut Analgesia, BP38, Clermont-Ferrand, France
| | - Xavier Moisset
- Université Clermont Auvergne, CHU de Clermont-Ferrand, Inserm, Neuro-Dol, Faculté de Médecine, Institut Analgesia, BP38, Clermont-Ferrand, France
| | - Laurence Daulhac
- Université Clermont Auvergne, Inserm, Neuro-Dol, Faculté de Pharmacie, Faculté de Médecine, Institut Analgesia, BP38, Clermont-Ferrand, France
| | - Alain Eschalier
- Université Clermont Auvergne, Inserm, Neuro-Dol, Faculté de Pharmacie, Faculté de Médecine, Institut Analgesia, BP38, Clermont-Ferrand, France
| | - Mélina Bégou
- Université Clermont Auvergne, Inserm, Neuro-Dol, Faculté de Pharmacie, Faculté de Médecine, Institut Analgesia, BP38, Clermont-Ferrand, France
- *Correspondence: Mélina Bégou
| |
Collapse
|
24
|
Kwon O. Pathophysiology of neuropathic pain. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2021. [DOI: 10.5124/jkma.2021.64.7.468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Background: Neuropathic pain is notoriously difficult to manage properly, not only because of its varied nature and the absence of objective diagnostic tools but also because of extensive reciprocal neuronal interactive pathogenic mechanism from the molecular level to patient’s own psychophysical characteristics. This paper briefly reviews the pathophysiology of neuropathic pain to the level of clinicians’ interest and its potential in clinical practiceCurrent Concepts: Recent research progress now allows us to obtain a bird view of neuropathic pain pathophysiology: peripheral and central sensitization. For peripheral sensitization, a local inflammatory milieu of the injured nerve primarily drives sequential phenotypic changes, which are critical and shared by both neuropathic and inflammatory pain. Central sensitization is led either by the hyperexcitability of the second-order afferent neuron itself or loss of physiological inhibitory control of the transmission of pain signal to the higher nervous system. Peripheral and central sensitization work synergistically but can also introduce neuropathic pain alone.Discussion and Conclusion: The cause of neuropathic pain is diverse, and understanding of its pathophysiology is still insufficient to realize a mechanism-based approach to clinical phenotypes or therapeutic applications. In dealing with chronic neuropathic pain, it is highly desirable to assess key aspects of a patient’s pain based on a plausible mechanism and select the best management method accordingly.
Collapse
|
25
|
Haroutounian S, Arendt-Nielsen L, Belton J, Blyth FM, Degenhardt L, Forti MD, Eccleston C, Finn DP, Finnerup NB, Fisher E, Fogarty AE, Gilron I, Hohmann AG, Kalso E, Krane E, Mohiuddin M, Moore RA, Rowbotham M, Soliman N, Wallace M, Zinboonyahgoon N, Rice ASC. International Association for the Study of Pain Presidential Task Force on Cannabis and Cannabinoid Analgesia: research agenda on the use of cannabinoids, cannabis, and cannabis-based medicines for pain management. Pain 2021; 162:S117-S124. [PMID: 34138827 PMCID: PMC8855877 DOI: 10.1097/j.pain.0000000000002266] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/10/2021] [Indexed: 01/08/2023]
Abstract
ABSTRACT The President of the International Association for the Study of Pain established a task force on cannabis and cannabinoid analgesia to systematically examine the evidence on (1) analgesic pharmacology of cannabinoids and preclinical evidence on their efficacy in animal models of injury-related or pathological persistent pain; (2) the clinical efficacy of cannabis, cannabinoids, and cannabis-based medicines for pain; (3) harms related to long-term use of cannabinoids; as well as (4) societal issues and policy implications related to the use of these compounds for pain management. Here, we summarize key knowledge gaps identified in the task force outputs and propose a research agenda for generating high-quality evidence on the topic. The systematic assessment of preclinical and clinical literature identified gaps in rigor of study design and reporting across the translational spectrum. We provide recommendations to improve the quality, rigor, transparency, and reproducibility of preclinical and clinical research on cannabis and cannabinoids for pain, as well as for the conduct of systematic reviews on the topic. Gaps related to comprehensive understanding of the endocannabinoid system and cannabinoid pharmacology, including pharmacokinetics and drug formulation aspects, are discussed. We outline key areas where high-quality clinical trials with cannabinoids are needed. Remaining important questions about long-term and short-term safety of cannabis and cannabinoids are emphasized. Finally, regulatory, societal, and policy challenges associated with medicinal and nonmedicinal use of cannabis are highlighted, with recommendations for improving patient safety and reducing societal harms in the context of pain management.
Collapse
Affiliation(s)
- Simon Haroutounian
- Division of Clinical and Translational Research and Washington University Pain Center. Department of Anesthesiology, Washington University School of Medicine. St Louis, MO, USA
| | - Lars Arendt-Nielsen
- Center for Neuroplasticity and Pain (CNAP) and Center for Sensory-Motor Interaction (SMI), Department of Health Science and Technology, School of Medicine, Aalborg University, Aalborg, Denmark
| | - Joletta Belton
- Endless Possibilities Initiative, Fraser, CO, USA; Global Alliance of Pain Patient Advocates (GAPPA) Presidential Task Force
| | - Fiona M. Blyth
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
| | - Louisa Degenhardt
- National Drug and Alcohol Research Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Marta Di Forti
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK. National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London, UK. South London and Maudsley NHS Mental Health Foundation Trust, London, UK
| | - Christopher Eccleston
- Centre for Pain Research. The University of Bath, Bath, UK, & Department of Clinical and Health Psychology, The University of Ghent, Belgium
| | - David P. Finn
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, Human Biology Building, National University of Ireland Galway, University Road, Galway, Ireland
| | - Nanna B Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Emma Fisher
- Centre for Pain Research. The University of Bath, Bath, UK
| | - Alexandra E. Fogarty
- Department of Neurology, Division of Physical Medicine & Rehabilitation, Washington University School of Medicine. St Louis, MO, USA
| | - Ian Gilron
- Departments of Anesthesiology & Perioperative Medicine and Biomedical & Molecular Sciences, Kingston Health Sciences Centre and Queen’s University; Centre for Neuroscience Studies, Queen’s University; School of Policy Studies, Queen’s University, Kingston, Canada
| | - Andrea G. Hohmann
- Department of Psychological and Brain Sciences, Program in Neuroscience, Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA
| | - Eija Kalso
- Department of Pharmacology and SleepWell Research Programme, University of Helsinki; Department of Anaesthesiology, Intensive Care and Pain Medicine, Helsinki University Hospital
| | - Elliot Krane
- Departments of Anesthesiology, Perioperative, and Pain Medicine, & Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Mohammed Mohiuddin
- Department of Anesthesiology & Perioperative Medicine and, Queen’s University, Kingston, Canada
| | | | - Michael Rowbotham
- Department of Anesthesia, University of California, San Francisco, California, USA
| | - Nadia Soliman
- Pain Research, Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, UK
| | - Mark Wallace
- Division of Pain Medicine, Department of Anesthesiology, University of California San Diego
| | | | - Andrew SC Rice
- Pain Research, Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, UK
| |
Collapse
|
26
|
Finn DP, Haroutounian S, Hohmann AG, Krane E, Soliman N, Rice ASC. Cannabinoids, the endocannabinoid system, and pain: a review of preclinical studies. Pain 2021; 162:S5-S25. [PMID: 33729211 PMCID: PMC8819673 DOI: 10.1097/j.pain.0000000000002268] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/10/2021] [Indexed: 12/13/2022]
Abstract
ABSTRACT This narrative review represents an output from the International Association for the Study of Pain's global task force on the use of cannabis, cannabinoids, and cannabis-based medicines for pain management, informed by our companion systematic review and meta-analysis of preclinical studies in this area. Our aims in this review are (1) to describe the value of studying cannabinoids and endogenous cannabinoid (endocannabinoid) system modulators in preclinical/animal models of pain; (2) to discuss both pain-related efficacy and additional pain-relevant effects (adverse and beneficial) of cannabinoids and endocannabinoid system modulators as they pertain to animal models of pathological or injury-related persistent pain; and (3) to identify important directions for future research. In service of these goals, this review (1) provides an overview of the endocannabinoid system and the pharmacology of cannabinoids and endocannabinoid system modulators, with specific relevance to animal models of pathological or injury-related persistent pain; (2) describes pharmacokinetics of cannabinoids in rodents and humans; and (3) highlights differences and discrepancies between preclinical and clinical studies in this area. Preclinical (rodent) models have advanced our understanding of the underlying sites and mechanisms of action of cannabinoids and the endocannabinoid system in suppressing nociceptive signaling and behaviors. We conclude that substantial evidence from animal models supports the contention that cannabinoids and endocannabinoid system modulators hold considerable promise for analgesic drug development, although the challenge of translating this knowledge into clinically useful medicines is not to be underestimated.
Collapse
Affiliation(s)
- David P Finn
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, Human Biology Building, National University of Ireland Galway, University Road, Galway, Ireland
| | - Simon Haroutounian
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Andrea G Hohmann
- Psychological and Brain Sciences, Program in Neuroscience, and Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA
| | - Elliot Krane
- Departments of Anesthesiology, Perioperative, and Pain Medicine, & Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Nadia Soliman
- Pain Research, Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, UK
| | - Andrew SC Rice
- Pain Research, Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, UK
| |
Collapse
|
27
|
Soliman N, Haroutounian S, Hohmann AG, Krane E, Liao J, Macleod M, Segelcke D, Sena C, Thomas J, Vollert J, Wever K, Alaverdyan H, Barakat A, Barthlow T, Bozer ALH, Davidson A, Diaz-delCastillo M, Dolgorukova A, Ferdousi MI, Healy C, Hong S, Hopkins M, James A, Leake HB, Malewicz NM, Mansfield M, Mardon AK, Mattimoe D, McLoone DP, Noes-Holt G, Pogatzki-Zahn EM, Power E, Pradier B, Romanos-Sirakis E, Segelcke A, Vinagre R, Yanes JA, Zhang J, Zhang XY, Finn DP, Rice AS. Systematic review and meta-analysis of cannabinoids, cannabis-based medicines, and endocannabinoid system modulators tested for antinociceptive effects in animal models of injury-related or pathological persistent pain. Pain 2021; 162:S26-S44. [PMID: 33729209 PMCID: PMC8216112 DOI: 10.1097/j.pain.0000000000002269] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/12/2021] [Accepted: 03/12/2021] [Indexed: 12/11/2022]
Abstract
ABSTRACT We report a systematic review and meta-analysis of studies that assessed the antinociceptive efficacy of cannabinoids, cannabis-based medicines, and endocannabinoid system modulators on pain-associated behavioural outcomes in animal models of pathological or injury-related persistent pain. In April 2019, we systematically searched 3 online databases and used crowd science and machine learning to identify studies for inclusion. We calculated a standardised mean difference effect size for each comparison and performed a random-effects meta-analysis. We assessed the impact of study design characteristics and reporting of mitigations to reduce the risk of bias. We meta-analysed 374 studies in which 171 interventions were assessed for antinociceptive efficacy in rodent models of pathological or injury-related pain. Most experiments were conducted in male animals (86%). Antinociceptive efficacy was most frequently measured by attenuation of hypersensitivity to evoked limb withdrawal. Selective cannabinoid type 1, cannabinoid type 2, nonselective cannabinoid receptor agonists (including delta-9-tetrahydrocannabinol) and peroxisome proliferator-activated receptor-alpha agonists (predominantly palmitoylethanolamide) significantly attenuated pain-associated behaviours in a broad range of inflammatory and neuropathic pain models. Fatty acid amide hydrolase inhibitors, monoacylglycerol lipase inhibitors, and cannabidiol significantly attenuated pain-associated behaviours in neuropathic pain models but yielded mixed results in inflammatory pain models. The reporting of criteria to reduce the risk of bias was low; therefore, the studies have an unclear risk of bias. The value of future studies could be enhanced by improving the reporting of methodological criteria, the clinical relevance of the models, and behavioural assessments. Notwithstanding, the evidence supports the hypothesis of cannabinoid-induced analgesia.
Collapse
Affiliation(s)
- Nadia Soliman
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Simon Haroutounian
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Andrea G. Hohmann
- Department of Psychological and Brain Sciences, Program in Neuroscience and Gill Center for Biomolecular Science, Bloomington, IN, United States
| | - Elliot Krane
- Departments of Anesthesiology, Perioperative, and Pain Medicine, & Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Jing Liao
- CAMARADES, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Malcolm Macleod
- CAMARADES, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Daniel Segelcke
- Department of Anesthesiology, Intensive Care and Pain Medicine University Hospital Muenster, Muenster, Germany
| | - Christopher Sena
- CAMARADES, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - James Thomas
- EPPI-Centre, University College London, London, United Kingdom
| | - Jan Vollert
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Kimberley Wever
- SYRCLE at Central Animal Laboratory, Radbound University Medical Center, Nijmegen, the Netherlands
| | - Harutyun Alaverdyan
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Ahmed Barakat
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Asyut, Egypt
| | - Tyler Barthlow
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amber L. Harris Bozer
- Department of Psychological Sciences, Tarleton State University, Stephenville, TX, United States
| | | | - Marta Diaz-delCastillo
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Antonina Dolgorukova
- Valdman Institute of Pharmacology, Pavlov First Saint Petersburg State Medical University, Saint Petersburg, Russia
| | - Mehnaz I. Ferdousi
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, Human Biology Building, National University of Ireland Galway, Galway, Ireland
| | - Catherine Healy
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, Human Biology Building, National University of Ireland Galway, Galway, Ireland
| | - Simon Hong
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Mary Hopkins
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, Human Biology Building, National University of Ireland Galway, Galway, Ireland
| | - Arul James
- Leicester General Hospital, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Hayley B. Leake
- IIMPACT in Health, University of South Australia, Adelaide, South Australia, Australia
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, New South Wales, Australia
| | - Nathalie M. Malewicz
- Department of Anaesthesiology, Intensive Care Medicine and Pain Management, Medical Faculty of Ruhr-University Bochum, BG University Hospital Bergmannsheil gGmbH, Bochum, Germany
| | - Michael Mansfield
- Department of Allied Health Sciences, Institute of Health and Social Care, Pain Research Cluster, Ageing, Acute and Long Term Conditions Research Group, London South Bank University, London, United Kingdom
| | - Amelia K. Mardon
- IIMPACT in Health, University of South Australia, Adelaide, South Australia, Australia
| | - Darragh Mattimoe
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, Human Biology Building, National University of Ireland Galway, Galway, Ireland
| | - Daniel P. McLoone
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, Human Biology Building, National University of Ireland Galway, Galway, Ireland
| | - Gith Noes-Holt
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Esther M. Pogatzki-Zahn
- Department of Anesthesiology, Intensive Care and Pain Medicine University Hospital Muenster, Muenster, Germany
| | - Emer Power
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, Human Biology Building, National University of Ireland Galway, Galway, Ireland
| | - Bruno Pradier
- Department of Anesthesiology, Intensive Care and Pain Medicine University Hospital Muenster, Muenster, Germany
| | - Eleny Romanos-Sirakis
- Staten Island University Hospital Northwell Health, Staten Island, NY, United States
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | | | - Rafael Vinagre
- Visiting Scholar, Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Julio A. Yanes
- Department of Psychological Sciences, Auburn University, Auburn, AL, United States
| | - Jingwen Zhang
- King's College London GKT School of Medical Education, King's College London, London, United Kingdom
| | - Xue Ying Zhang
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - David P. Finn
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, Human Biology Building, National University of Ireland Galway, Galway, Ireland
| | - Andrew S.C. Rice
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| |
Collapse
|
28
|
Hore ZL, Villa-Hernandez S, Denk F. Probing the peripheral immune response in mouse models of oxaliplatin-induced peripheral neuropathy highlights their limited translatability. Wellcome Open Res 2021; 6:68. [PMID: 34250264 PMCID: PMC8243229 DOI: 10.12688/wellcomeopenres.16635.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Chemotherapy-induced peripheral neuropathy (CIPN) is a disabling side effect of various chemotherapeutic agents, including oxaliplatin. It is highly prevalent amongst cancer patients, causing sensory abnormalities and pain. Unfortunately, as the underlying mechanisms remain poorly understood, effective therapeutics are lacking. Neuro-immune interactions have been highlighted as potential contributors to the development and maintenance of CIPN, however, whether this is the case in oxaliplatin-induced peripheral neuropathy (OIPN) is yet to be fully established. Methods: In this study we used flow cytometry to examine the peripheral immune response of male C57BL/6 mice following both single and repeated oxaliplatin administration. In animals exposed to repeated dosing, we also undertook mechanical and thermal behavioural assays to investigate how oxaliplatin alters phenotype, and conducted RT-qPCR experiments on bone marrow derived macrophages in order to further inspect the effects of oxaliplatin on immune cells. Results: In contrast to other reports, we failed to observe substantial changes in overall leukocyte, lymphocyte or myeloid cell numbers in dorsal root ganglia, sciatic nerves or inguinal lymph nodes. We did however note subtle, tissue-dependant alterations in several myeloid subpopulations following repeated dosing. These included a significant reduction in MHCII antigen presenting cells in the sciatic nerve and an increase in infiltrating cell types into the inguinal lymph nodes. Though repeated oxaliplatin administration had a systemic effect, we were unable to detect a pain-like behavioural phenotype in response to either cold or mechanical stimuli. Consequently, we cannot comment on whether the observed myeloid changes are associated with OIPN. Conclusions: Our discussion puts these results into the wider context of the field, advocating for greater transparency in reporting, alignment in experimental design and the introduction of more clinically relevant models. Only through joint concerted effort can we hope to increase our understanding of the underlying mechanisms of CIPN, including any immune contributions.
Collapse
Affiliation(s)
- Zoe Lee Hore
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| | - Sara Villa-Hernandez
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| | - Franziska Denk
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| |
Collapse
|
29
|
A randomized, controlled trial of a β2-agonist in painful polyneuropathy. Pain 2021; 162:1364-1373. [PMID: 33181580 DOI: 10.1097/j.pain.0000000000002140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023]
Abstract
ABSTRACT Experimental data have suggested that in neuropathic pain, tricyclic antidepressants may work solely through a β2-agonist action. The aim of this study was to test if the β2-agonist terbutaline relieves painful polyneuropathy. The study was a randomized, double-blind, placebo-controlled and active-controlled, 3-way, cross-over trial among patients with painful polyneuropathy. The treatment periods were of 5 weeks' duration and were preceded by 1 week for washout and 1 week for baseline observations. The patients received terbutaline (5-15 mg), imipramine (30-150 mg), or placebo in a random order. Drug doses depended on age and metabolizer status. The change in total pain recorded from ratings in diaries (numeric rating scale [NRS] 0-10) was the primary outcome, and the change in rating of specific pain symptoms (NRS 0-10), patient global impression of change, and sleep disturbance were secondary outcomes. Forty-seven patients were randomized. The median score for total pain changed from NRS 6.4 to 6.1 from baseline to week 5 on terbutaline with an average effect during the treatment period as compared with placebo of 0.13 (95% confidence interval -0.12 to 0.38, P = 0.32). The median score for total pain on imipramine changed from NRS 6.6 to 4.8 with an average effect as compared with placebo of -1.17 (95% confidence interval -1.42 to -0.92, P < 0.001). Secondary outcomes were also unaltered by terbutaline but improved by imipramine. The β2-agonist terbutaline has no effect in painful polyneuropathy. β2-agonism seems not to be an important mechanism of action of tricyclic antidepressants in neuropathic pain.
Collapse
|
30
|
Abstract
The role of immune mediators, including proinflammatory cytokines in chemotherapy-induced peripheral neuropathy (CIPN), remains unclear. Here, we studied the contribution of interleukin-20 (IL-20) to the development of paclitaxel-induced peripheral neuropathy. Increased serum levels of IL-20 in cancer patients with chemotherapy were accompanied by increased CIPN risk. In mouse models, proinflammatory IL-20 levels in serum and dorsal root ganglia fluctuated with paclitaxel treatment. Blocking IL-20 with the neutralizing antibody or genetic deletion of its receptors prevented CIPN, alleviated peripheral nerve damage, and dampened inflammatory responses, including macrophage infiltration and cytokine release. Mechanistically, paclitaxel upregulated IL-20 through dysregulated Ca homeostasis, which augmented chemotherapy-induced neurotoxicity. Importantly, IL-20 suppression did not alter paclitaxel efficacy on cancer treatment both in vitro and in vivo. Together, targeting IL-20 ameliorates paclitaxel-induced peripheral neuropathy by suppressing neuroinflammation and restoring Ca homeostasis. Therefore, the anti-IL-20 monoclonal antibody is a promising therapeutic for the prevention and treatment of paclitaxel-induced neuropathy.
Collapse
|
31
|
Fan W, Sdrulla AD. Differential modulation of excitatory and inhibitory populations of superficial dorsal horn neurons in lumbar spinal cord by Aβ-fiber electrical stimulation. Pain 2021; 161:1650-1660. [PMID: 32068665 DOI: 10.1097/j.pain.0000000000001836] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Activation of Aβ-fibers is fundamental to numerous analgesic therapies, yet its effects on dorsal horn neuronal activity remain unclear. We used multiphoton microscopy of the genetically encoded calcium indicator GCaMP6s to characterize the effects of Aβ-fiber electrical stimulation (Aβ-ES) on neural activity. Specifically, we quantified somatic responses evoked by C-fiber intensity stimulation before and after a 10-minute train of dorsal root Aβ-ES in superficial dorsal horn (SDH) neurons, in mouse lumbar spinal cord. Aβ-ES did not alter C-fiber-evoked activity when GCaMP6s was virally expressed in all neurons, in an intact lumbar spinal cord preparation. However, when we restricted the expression of GCaMP6s to excitatory or inhibitory populations, we observed that Aβ-ES modestly potentiated evoked activity of excitatory neurons and depressed that of inhibitory neurons. Aβ-ES had no significant effects in a slice preparation in either SDH population. A larger proportion of SDH neurons was activated by Aβ-ES when delivered at a root rostral or caudal to the segment where the imaging and C-fiber intensity stimulation occurred. Aβ-ES effects on excitatory and inhibitory populations depended on the root used. Our findings suggest that Aβ-ES differentially modulates lumbar spinal cord SDH populations in a cell type- and input-specific manner. Furthermore, they underscore the importance of the Aβ-ES delivery site, suggesting that Aβ stimulation at a segment adjacent to where the pain is may improve analgesic efficacy.
Collapse
Affiliation(s)
- Wei Fan
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| | | |
Collapse
|
32
|
Hoogendoorn A, Avery TD, Li J, Bursill C, Abell A, Grace PM. Emerging Therapeutic Applications for Fumarates. Trends Pharmacol Sci 2021; 42:239-254. [PMID: 33618840 PMCID: PMC7954891 DOI: 10.1016/j.tips.2021.01.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 02/08/2023]
Abstract
Fumarates are successfully used for the treatment of psoriasis and multiple sclerosis. Their antioxidative, immunomodulatory, and neuroprotective properties make fumarates attractive therapeutic candidates for other pathologies. The exact working mechanisms of fumarates are, however, not fully understood. Further elucidation of the mechanisms is required if these drugs are to be successfully repurposed for other diseases. Towards this, administration route, dosage, and treatment timing, frequency, and duration are important parameters to consider and optimize with clinical paradigms in mind. Here, we summarize the rapidly expanding literature on the pharmacokinetics and pharmacodynamics of fumarates, including a discussion on two recently FDA-approved fumarates VumerityTM and BafiertamTM. We review emerging applications of fumarates, focusing on neurological and cardiovascular diseases.
Collapse
Affiliation(s)
- Ayla Hoogendoorn
- Vascular and Heart Health, Life Long Health Theme, South Australian Health and Medical Research Institute, Adelaide, Australia; ARC Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Australia.
| | - Thomas D Avery
- ARC Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Australia; Institute for Photonics and Advanced Sensing & Department of Chemistry, The University of Adelaide, Australia
| | - Jiahe Li
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christina Bursill
- Vascular and Heart Health, Life Long Health Theme, South Australian Health and Medical Research Institute, Adelaide, Australia; ARC Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Australia; Faculty of Health and Medical Science, University of Adelaide, Adelaide, Australia
| | - Andrew Abell
- ARC Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Australia; Institute for Photonics and Advanced Sensing & Department of Chemistry, The University of Adelaide, Australia
| | - Peter M Grace
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
33
|
Hore ZL, Villa-Hernandez S, Denk F. Probing the peripheral immune response in mouse models of oxaliplatin-induced peripheral neuropathy highlights their limited translatability. Wellcome Open Res 2021; 6:68. [PMID: 34250264 PMCID: PMC8243229 DOI: 10.12688/wellcomeopenres.16635.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2021] [Indexed: 04/03/2024] Open
Abstract
Background: Chemotherapy-induced peripheral neuropathy (CIPN) is a disabling side effect of various chemotherapeutic agents, including oxaliplatin. It is highly prevalent amongst cancer patients, causing sensory abnormalities and pain. Unfortunately, as the underlying mechanisms remain poorly understood, effective therapeutics are lacking. Neuro-immune interactions have been highlighted as potential contributors to the development and maintenance of CIPN, however, whether this is the case in oxaliplatin-induced peripheral neuropathy (OIPN) is yet to be fully established. Methods: In this study we used flow cytometry to examine the peripheral immune response of male C57BL/6 mice following both single and repeated oxaliplatin administration. In animals exposed to repeated dosing, we also undertook mechanical and thermal behavioural assays to investigate how oxaliplatin alters phenotype, and conducted RT-qPCR experiments on bone marrow derived macrophages in order to further inspect the effects of oxaliplatin on immune cells. Results: In contrast to other reports, we failed to observe substantial changes in overall leukocyte, lymphocyte or myeloid cell numbers in dorsal root ganglia, sciatic nerves or inguinal lymph nodes. We did however note subtle, tissue-dependant alterations in several myeloid subpopulations following repeated dosing. These included a significant reduction in MHCII antigen presenting cells in the sciatic nerve and an increase in infiltrating cell types into the inguinal lymph nodes. Though repeated oxaliplatin administration had a systemic effect, we were unable to detect a pain-like behavioural phenotype in response to either cold or mechanical stimuli. Consequently, we cannot comment on whether the observed myeloid changes are associated with OIPN. Conclusions: Our discussion puts these results into the wider context of the field, advocating for greater transparency in reporting, alignment in experimental design and the introduction of more clinically relevant models. Only through joint concerted effort can we hope to increase our understanding of the underlying mechanisms of CIPN, including any immune contributions.
Collapse
Affiliation(s)
- Zoe Lee Hore
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| | - Sara Villa-Hernandez
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| | - Franziska Denk
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| |
Collapse
|
34
|
Fisher AS, Lanigan MT, Upton N, Lione LA. Preclinical Neuropathic Pain Assessment; the Importance of Translatability and Bidirectional Research. Front Pharmacol 2021; 11:614990. [PMID: 33628181 PMCID: PMC7897667 DOI: 10.3389/fphar.2020.614990] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/10/2020] [Indexed: 02/04/2023] Open
Abstract
For patients suffering with chronic neuropathic pain the need for suitable novel therapies is imperative. Over recent years a contributing factor for the lack of development of new analgesics for neuropathic pain has been the mismatch of primary neuropathic pain assessment endpoints in preclinical vs. clinical trials. Despite continuous forward translation failures across diverse mechanisms, reflexive quantitative sensory testing remains the primary assessment endpoint for neuropathic pain and analgesia in animals. Restricting preclinical evaluation of pain and analgesia to exclusively reflexive outcomes is over simplified and can be argued not clinically relevant due to the continued lack of forward translation and failures in the clinic. The key to developing new analgesic treatments for neuropathic pain therefore lies in the development of clinically relevant endpoints that can translate preclinical animal results to human clinical trials. In this review we discuss this mismatch of primary neuropathic pain assessment endpoints, together with clinical and preclinical evidence that supports how bidirectional research is helping to validate new clinically relevant neuropathic pain assessment endpoints. Ethological behavioral endpoints such as burrowing and facial grimacing and objective measures such as electroencephalography provide improved translatability potential together with currently used quantitative sensory testing endpoints. By tailoring objective and subjective measures of neuropathic pain the translatability of new medicines for patients suffering with neuropathic pain will hopefully be improved.
Collapse
Affiliation(s)
- Amy S. Fisher
- Transpharmation Ltd., The London Bioscience Innovation Centre, London, United Kingdom
| | - Michael T. Lanigan
- Transpharmation Ltd., The London Bioscience Innovation Centre, London, United Kingdom
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - Neil Upton
- Transpharmation Ltd., The London Bioscience Innovation Centre, London, United Kingdom
| | - Lisa A. Lione
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| |
Collapse
|
35
|
The influence of rat strain on the development of neuropathic pain and comorbid anxio-depressive behaviour after nerve injury. Sci Rep 2020; 10:20981. [PMID: 33262364 PMCID: PMC7708988 DOI: 10.1038/s41598-020-77640-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 11/13/2020] [Indexed: 12/16/2022] Open
Abstract
Back-translating the clinical manifestations of human disease burden into animal models is increasingly recognized as an important facet of preclinical drug discovery. We hypothesized that inbred rat strains possessing stress hyper-reactive-, depressive- or anxiety-like phenotypes may possess more translational value than common outbred strains for modeling neuropathic pain. Rats (inbred: LEW, WKY, F344/ICO and F344/DU, outbred: Crl:SD) were exposed to Spared Nerve Injury (SNI) and evaluated routinely for 6 months on behaviours related to pain (von Frey stimulation and CatWalk-gait analysis), anxiety (elevated plus maze, EPM) and depression (sucrose preference test, SPT). Markers of stress reactivity together with spinal/brain opioid receptor expression were also measured. All strains variously developed mechanical allodynia after SNI with the exception of stress-hyporesponsive LEW rats, despite all strains displaying similar functional gait-deficits after injury. However, affective changes reflective of anxiety- and depressive-like behaviour were only observed for F344/DU in the EPM, and for Crl:SD in SPT. Although differences in stress reactivity and opioid receptor expression occurred, overall they were relatively unaffected by SNI. Thus, anxio-depressive behaviours did not develop in all strains after nerve injury, and correlated only modestly with degree of pain sensitivity or with genetic predisposition to stress and/or affective disturbances.
Collapse
|
36
|
Abboud C, Duveau A, Bouali-Benazzouz R, Massé K, Mattar J, Brochoire L, Fossat P, Boué-Grabot E, Hleihel W, Landry M. Animal models of pain: Diversity and benefits. J Neurosci Methods 2020; 348:108997. [PMID: 33188801 DOI: 10.1016/j.jneumeth.2020.108997] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/03/2020] [Accepted: 11/08/2020] [Indexed: 12/15/2022]
Abstract
Chronic pain is a maladaptive neurological disease that remains a major health problem. A deepening of our knowledge on mechanisms that cause pain is a prerequisite to developing novel treatments. A large variety of animal models of pain has been developed that recapitulate the diverse symptoms of different pain pathologies. These models reproduce different pain phenotypes and remain necessary to examine the multidimensional aspects of pain and understand the cellular and molecular basis underlying pain conditions. In this review, we propose an overview of animal models, from simple organisms to rodents and non-human primates and the specific traits of pain pathologies they model. We present the main behavioral tests for assessing pain and investing the underpinning mechanisms of chronic pathological pain. The validity of animal models is analysed based on their ability to mimic human clinical diseases and to predict treatment outcomes. Refine characterization of pathological phenotypes also requires to consider pain globally using specific procedures dedicated to study emotional comorbidities of pain. We discuss the limitations of pain models when research findings fail to be translated from animal models to human clinics. But we also point to some recent successes in analgesic drug development that highlight strategies for improving the predictive validity of animal models of pain. Finally, we emphasize the importance of using assortments of preclinical pain models to identify pain subtype mechanisms, and to foster the development of better analgesics.
Collapse
Affiliation(s)
- Cynthia Abboud
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France; Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France; Faculty of Arts and Sciences, Holy Spirit University of Kaslik (USEK), Lebanon
| | - Alexia Duveau
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Rabia Bouali-Benazzouz
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Karine Massé
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Joseph Mattar
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Lebanon
| | - Louison Brochoire
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Pascal Fossat
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Eric Boué-Grabot
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Walid Hleihel
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Lebanon; Faculty of Arts and Sciences, Holy Spirit University of Kaslik (USEK), Lebanon
| | - Marc Landry
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France.
| |
Collapse
|
37
|
Fatty acid suppression of glial activation prevents central neuropathic pain after spinal cord injury. Pain 2020; 160:2724-2742. [PMID: 31365471 DOI: 10.1097/j.pain.0000000000001670] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
About half of patients with spinal cord injury (SCI) develop debilitating central neuropathic pain (CNP), with no effective treatments. Thus, effective, safe, and novel therapies are needed urgently. Previously, docosahexaenoic acid (DHA) was reported to confer neuroprotection in preclinical SCI models. However, its therapeutic potential on SCI-CNP remains to be elucidated. Here, we demonstrated for the first time that intravenous DHA administrations with 3-day intervals (250 nmol/kg; starting 30 minutes after injury and maintained for 6 weeks) effectively prevented SCI-CNP development in a clinically relevant rat contusion model. SCI-CNP was assessed by a novel sensory profiling approach combining evoked pain measures and pain-related ethologically relevant rodent behaviours (burrowing, thigmotaxis, and place/escape avoidance) to mimic those for measuring human (sensory, affective, cognitive, and spontaneous) pain. Strikingly, already established SCI-CNP could be abolished partially by similar DHA administrations, starting from the beginning of week 4 after injury and maintained for 4 weeks. At spinal (epicenter and L5 dorsal horns) and supraspinal (anterior cingulate cortex) levels, both treatment regimens potently suppressed microglial and astrocyte activation, which underpins SCI-CNP pathogenesis. Spinal microgliosis, a known hallmark associated with neuropathic pain behaviours, was reduced by DHA treatments. Finally, we revealed novel potential roles of peroxisome proliferator-activated and retinoid X receptors and docosahexaenoyl ethanolamide (DHA's metabolite) in mediating DHA's effects on microglial activation. Our findings, coupled with the excellent long-term clinical safety of DHA even in surgical and critically ill patients, suggest that systemic DHA treatment is a translatable, effective, safe, and novel approach for preventing and managing SCI-CNP.
Collapse
|
38
|
Cabañero D, Ramírez-López A, Drews E, Schmöle A, Otte DM, Wawrzczak-Bargiela A, Huerga Encabo H, Kummer S, Ferrer-Montiel A, Przewlocki R, Zimmer A, Maldonado R. Protective role of neuronal and lymphoid cannabinoid CB 2 receptors in neuropathic pain. eLife 2020; 9:55582. [PMID: 32687056 PMCID: PMC7384863 DOI: 10.7554/elife.55582] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 07/19/2020] [Indexed: 12/14/2022] Open
Abstract
Cannabinoid CB2 receptor (CB2) agonists are potential analgesics void of psychotropic effects. Peripheral immune cells, neurons and glia express CB2; however, the involvement of CB2 from these cells in neuropathic pain remains unresolved. We explored spontaneous neuropathic pain through on-demand self-administration of the selective CB2 agonist JWH133 in wild-type and knockout mice lacking CB2 in neurons, monocytes or constitutively. Operant self-administration reflected drug-taking to alleviate spontaneous pain, nociceptive and affective manifestations. While constitutive deletion of CB2 disrupted JWH133-taking behavior, this behavior was not modified in monocyte-specific CB2 knockouts and was increased in mice defective in neuronal CB2 knockouts suggestive of increased spontaneous pain. Interestingly, CB2-positive lymphocytes infiltrated the injured nerve and possible CB2transfer from immune cells to neurons was found. Lymphocyte CB2depletion also exacerbated JWH133 self-administration and inhibited antinociception. This work identifies a simultaneous activity of neuronal and lymphoid CB2that protects against spontaneous and evoked neuropathic pain.
Collapse
Affiliation(s)
- David Cabañero
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,Institute of Research, Development and Innovation in Healthcare Biotechnology of Elche (IDiBE), Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Angela Ramírez-López
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Eva Drews
- Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | - Anne Schmöle
- Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | - David M Otte
- Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | - Agnieszka Wawrzczak-Bargiela
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Hector Huerga Encabo
- Immunology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Sami Kummer
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Antonio Ferrer-Montiel
- Institute of Research, Development and Innovation in Healthcare Biotechnology of Elche (IDiBE), Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Ryszard Przewlocki
- Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Andreas Zimmer
- Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| |
Collapse
|
39
|
Abstract
Neuropathic pain caused by a lesion or disease of the somatosensory nervous system is a common chronic pain condition with major impact on quality of life. Examples include trigeminal neuralgia, painful polyneuropathy, postherpetic neuralgia, and central poststroke pain. Most patients complain of an ongoing or intermittent spontaneous pain of, for example, burning, pricking, squeezing quality, which may be accompanied by evoked pain, particular to light touch and cold. Ectopic activity in, for example, nerve-end neuroma, compressed nerves or nerve roots, dorsal root ganglia, and the thalamus may in different conditions underlie the spontaneous pain. Evoked pain may spread to neighboring areas, and the underlying pathophysiology involves peripheral and central sensitization. Maladaptive structural changes and a number of cell-cell interactions and molecular signaling underlie the sensitization of nociceptive pathways. These include alteration in ion channels, activation of immune cells, glial-derived mediators, and epigenetic regulation. The major classes of therapeutics include drugs acting on α2δ subunits of calcium channels, sodium channels, and descending modulatory inhibitory pathways.
Collapse
Affiliation(s)
- Nanna Brix Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Rohini Kuner
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Troels Staehelin Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
40
|
The emergence of animal models of chronic pain and logistical and methodological issues concerning their use. J Neural Transm (Vienna) 2019; 127:393-406. [DOI: 10.1007/s00702-019-02103-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022]
|
41
|
Colvin LA, Rice ASC. Progress in pain medicine: where are we now? Br J Anaesth 2019; 123:e173-e176. [PMID: 31174848 PMCID: PMC6676231 DOI: 10.1016/j.bja.2019.04.051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 12/18/2022] Open
Affiliation(s)
- Lesley A Colvin
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK.
| | - Andrew S C Rice
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, UK
| |
Collapse
|
42
|
Obara I, Telezhkin V, Alrashdi I, Chazot PL. Histamine, histamine receptors, and neuropathic pain relief. Br J Pharmacol 2019; 177:580-599. [PMID: 31046146 PMCID: PMC7012972 DOI: 10.1111/bph.14696] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 02/21/2019] [Accepted: 04/18/2019] [Indexed: 01/06/2023] Open
Abstract
Histamine, acting via distinct histamine H1, H2, H3, and H4 receptors, regulates various physiological and pathological processes, including pain. In the last two decades, there has been a particular increase in evidence to support the involvement of H3 receptor and H4 receptor in the modulation of neuropathic pain, which remains challenging in terms of management. However, recent data show contrasting effects on neuropathic pain due to multiple factors that determine the pharmacological responses of histamine receptors and their underlying signal transduction properties (e.g., localization on either the presynaptic or postsynaptic neuronal membranes). This review summarizes the most recent findings on the role of histamine and the effects mediated by the four histamine receptors in response to the various stimuli associated with and promoting neuropathic pain. We particularly focus on mechanisms underlying histamine‐mediated analgesia, as we aim to clarify the analgesic potential of histamine receptor ligands in neuropathic pain. Linked Articles This article is part of a themed section on New Uses for 21st Century. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.3/issuetoc
Collapse
Affiliation(s)
- Ilona Obara
- School of Pharmacy, Newcastle University, Newcastle upon Tyne, UK.,Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Vsevolod Telezhkin
- School of Dental Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Ibrahim Alrashdi
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Paul L Chazot
- Department of Biosciences, Durham University, Durham, UK
| |
Collapse
|
43
|
Currie GL, Angel-Scott HN, Colvin L, Cramond F, Hair K, Khandoker L, Liao J, Macleod M, McCann SK, Morland R, Sherratt N, Stewart R, Tanriver-Ayder E, Thomas J, Wang Q, Wodarski R, Xiong R, Rice ASC, Sena ES. Animal models of chemotherapy-induced peripheral neuropathy: A machine-assisted systematic review and meta-analysis. PLoS Biol 2019; 17:e3000243. [PMID: 31107871 PMCID: PMC6544332 DOI: 10.1371/journal.pbio.3000243] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 05/31/2019] [Accepted: 04/08/2019] [Indexed: 12/25/2022] Open
Abstract
We report a systematic review and meta-analysis of research using animal models of chemotherapy-induced peripheral neuropathy (CIPN). We systematically searched 5 online databases in September 2012 and updated the search in November 2015 using machine learning and text mining to reduce the screening for inclusion workload and improve accuracy. For each comparison, we calculated a standardised mean difference (SMD) effect size, and then combined effects in a random-effects meta-analysis. We assessed the impact of study design factors and reporting of measures to reduce risks of bias. We present power analyses for the most frequently reported behavioural tests; 337 publications were included. Most studies (84%) used male animals only. The most frequently reported outcome measure was evoked limb withdrawal in response to mechanical monofilaments. There was modest reporting of measures to reduce risks of bias. The number of animals required to obtain 80% power with a significance level of 0.05 varied substantially across behavioural tests. In this comprehensive summary of the use of animal models of CIPN, we have identified areas in which the value of preclinical CIPN studies might be increased. Using both sexes of animals in the modelling of CIPN, ensuring that outcome measures align with those most relevant in the clinic, and the animal's pain contextualised ethology will likely improve external validity. Measures to reduce risk of bias should be employed to increase the internal validity of studies. Different outcome measures have different statistical power, and this can refine our approaches in the modelling of CIPN.
Collapse
Affiliation(s)
- Gillian L. Currie
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Helena N. Angel-Scott
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Lesley Colvin
- Department of Anaesthesia, Critical Care & Pain, University of Edinburgh, Edinburgh, United Kingdom
- Division of Population Health and Genomics, University of Dundee, Dundee, United Kingdom
| | - Fala Cramond
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Kaitlyn Hair
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Laila Khandoker
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Jing Liao
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Malcolm Macleod
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Sarah K. McCann
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Rosie Morland
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Nicki Sherratt
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Robert Stewart
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Ezgi Tanriver-Ayder
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - James Thomas
- EPPI-Centre, University College London, London, United Kingdom
| | - Qianying Wang
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Rachel Wodarski
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Ran Xiong
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Andrew S. C. Rice
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Emily S. Sena
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
44
|
|
45
|
Patel R, Kucharczyk M, Montagut‐Bordas C, Lockwood S, Dickenson AH. Neuropathy following spinal nerve injury shares features with the irritable nociceptor phenotype: A back-translational study of oxcarbazepine. Eur J Pain 2019; 23:183-197. [PMID: 30091265 PMCID: PMC6396087 DOI: 10.1002/ejp.1300] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/24/2018] [Accepted: 07/31/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND The term 'irritable nociceptor' was coined to describe neuropathic patients characterized by evoked hypersensitivity and preservation of primary afferent fibres. Oxcarbazepine is largely ineffectual in an overall patient population, but has clear efficacy in a subgroup with the irritable nociceptor profile. We examine whether neuropathy in rats induced by spinal nerve injury shares overlapping pharmacological sensitivity with the irritable nociceptor phenotype using drugs that target sodium channels. METHODS In vivo electrophysiology was performed in anaesthetized spinal nerve ligated (SNL) and sham-operated rats to record from wide dynamic range (WDR) neurones in the ventral posterolateral thalamus (VPL) and dorsal horn. RESULTS In neuropathic rats, spontaneous activity in the VPL was substantially attenuated by spinal lidocaine, an effect that was absent in sham rats. The former measure was in part dependent on ongoing peripheral activity as intraplantar lidocaine also reduced aberrant spontaneous thalamic firing. Systemic oxcarbazepine had no effect on wind-up of dorsal horn neurones in sham and SNL rats. However, in SNL rats, oxcarbazepine markedly inhibited punctate mechanical-, dynamic brush- and cold-evoked neuronal responses in the VPL and dorsal horn, with minimal effects on heat-evoked responses. In addition, oxcarbazepine inhibited spontaneous activity in the VPL. Intraplantar injection of the active metabolite licarbazepine replicated the effects of systemic oxcarbazepine, supporting a peripheral locus of action. CONCLUSIONS We provide evidence that ongoing activity in primary afferent fibres drives spontaneous thalamic firing after spinal nerve injury and that oxcarbazepine through a peripheral mechanism exhibits modality-selective inhibitory effects on sensory neuronal processing. SIGNIFICANCE The inhibitory effects of lidocaine and oxcarbazepine in this rat model of neuropathy resemble the clinical observations in the irritable nociceptor patient subgroup and support a mechanism-based rationale for bench-to-bedside translation when screening novel drugs.
Collapse
Affiliation(s)
- Ryan Patel
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| | - Mateusz Kucharczyk
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| | | | - Stevie Lockwood
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| | - Anthony H. Dickenson
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| |
Collapse
|
46
|
Getting a handle on Ca V2.2 (N-type) voltage-gated Ca 2+ channels. Proc Natl Acad Sci U S A 2018; 115:12848-12850. [PMID: 30538200 DOI: 10.1073/pnas.1818608115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
47
|
Reply. Pain 2018; 159:1428-1429. [PMID: 29916962 DOI: 10.1097/j.pain.0000000000001249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
48
|
Wodarski R, Bagdas D, Paris JJ, Pheby T, Toma W, Xu R, Damaj MI, Knapp PE, Rice AS, Hauser KF. Reduced intraepidermal nerve fibre density, glial activation, and sensory changes in HIV type-1 Tat-expressing female mice: involvement of Tat during early stages of HIV-associated painful sensory neuropathy. Pain Rep 2018; 3:e654. [PMID: 29922746 PMCID: PMC5999412 DOI: 10.1097/pr9.0000000000000654] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/19/2018] [Accepted: 03/17/2018] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION HIV infection is associated with chronic pain states, including sensory neuropathy, which affects greater than 40% of patients. OBJECTIVES AND METHODS To determine the impact of HIV-Tat induction on nociceptive behaviour in female mice conditionally expressing HIV Tat1-86 protein through a doxycycline (DOX)-driven glial fibrillary acidic protein promoter, intraepidermal nerve fibre density and immune cell activation in the dorsal root ganglion (DRG) and spinal cord were assessed by immunohistochemistry. Mice were assessed for mechanical and thermal sensitivity for 9 weeks using von-Frey and Hargreaves tests. RESULTS Intraepidermal nerve fibre density was significantly reduced after 6 weeks of Tat induction, similar to sensory neuropathy seen in clinical HIV infection. Tat induction through DOX caused a significant reduction in paw withdrawal thresholds in a time-dependent manner starting the 4th week after Tat induction. No changes in paw withdrawal latencies were seen in Tat(-) control mice lacking the tat transgene. Although reductions in paw withdrawal thresholds increased throughout the study, no significant change in spontaneous motor activity was observed. Spinal cord (cervical and lumbar), DRG, and hind paw skin were collected at 8 days and 6 weeks after Tat induction. HIV-Tat mRNA expression was significantly increased in lumbar DRG and skin samples 8 days after DOX treatment. Tat induced a significant increase in the number of Iba-1 positive cells at 6 weeks, but not after 8 days, of exposure. No differences in glial fibrillary acidic protein immunoreactivity were observed. CONCLUSION These results suggest that Tat protein contributes to painful HIV-related sensory neuropathy during the initial stages of the pathogenesis.
Collapse
Affiliation(s)
- Rachel Wodarski
- Pain Research Group, Department of Surgery and Cancer, Imperial College, Chelsea and Westminster Hospital Campus, London, United Kingdom
| | - Deniz Bagdas
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Jason J. Paris
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
- Department of BioMolecular Sciences, University of Mississippi, University, MS, USA
| | - Tim Pheby
- Pain Research Group, Department of Surgery and Cancer, Imperial College, Chelsea and Westminster Hospital Campus, London, United Kingdom
| | - Wisam Toma
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Ruqiang Xu
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - M. Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Pamela E. Knapp
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Andrew S.C. Rice
- Pain Research Group, Department of Surgery and Cancer, Imperial College, Chelsea and Westminster Hospital Campus, London, United Kingdom
| | - Kurt F. Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|