1
|
Patel KV, Gadotti VM, Garcia-Caballero A, Antunes FTT, Ali MY, Zamponi GW, Derksen DJ. Development of Tetrahydroquinoline-Based Inhibitors for Chronic Pain. ACS Chem Neurosci 2024. [PMID: 39377454 DOI: 10.1021/acschemneuro.4c00316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024] Open
Abstract
Chronic pain affects a substantial portion of the population, posing a significant health challenge. Current treatments often come with limitations and side effects, necessitating novel therapeutic approaches. Our study focuses on disrupting the Cav3.2-USP5 interaction as a strategy for chronic pain management. Through structure-activity relationship studies of a tetrahydroquinoline (THQ) scaffold, we identified a family of lead molecules that demonstrated potent inhibition of the Cav3.2-USP5 interaction. In vitro pharmacokinetic assessments and in vivo studies support the efficacy and drug-like properties of the lead compounds in mouse models of acute and chronic pain. Dependence on the Cav3.2 channels was validated in Cav3.2 null mice, consistent with the proposed mode of action of these small molecules. These findings provide a novel chronic pain treatment strategy, highlighting the potential of these small molecules for further development.
Collapse
Affiliation(s)
- Ketul V Patel
- Department of Chemistry, University of Calgary, Calgary T2N 1N4, Alberta, Canada
- Zymedyne Therapeutics, Calgary T2N 4G4, Alberta, Canada
| | - Vinicius M Gadotti
- Department of Clinical Neurosciences, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
| | - Agustin Garcia-Caballero
- Department of Clinical Neurosciences, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
| | - Flavia T T Antunes
- Department of Clinical Neurosciences, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
| | - Md Yousof Ali
- Department of Clinical Neurosciences, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
- Zymedyne Therapeutics, Calgary T2N 4G4, Alberta, Canada
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
| | - Darren J Derksen
- Department of Chemistry, University of Calgary, Calgary T2N 1N4, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
| |
Collapse
|
2
|
de Melo Cardoso M, Scussel R, da Silva Abel J, Pereira FO, Cruz LA, da Costa Constante F, De Pieri E, Abelaira HM, Ferreira J, Gomez MV, Rigo FK, Machado-de-Ávila RA. Intravenous administration of recombinant Phα1β: Antinociceptive properties and morphine tolerance reversal in a cancer-associated pain model. Toxicon 2024; 243:107717. [PMID: 38614245 DOI: 10.1016/j.toxicon.2024.107717] [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: 01/10/2024] [Revised: 03/12/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Cancer-related pain is considered one of the most prevalent symptoms for those affected by cancer, significantly influencing quality of life and treatment outcomes. Morphine is currently employed for analgesic treatment in this case, however, chronic use of this opioid is limited by the development of analgesic tolerance and adverse effects, such as digestive and neurological disorders. Alternative therapies, such as ion channel blockade, are explored. The toxin Phα1β has demonstrated efficacy in blocking calcium channels, making it a potential candidate for alleviating cancer-related pain. This study aims to assess the antinociceptive effects resulting from intravenous administration of the recombinant form of Phα1β (r-Phα1β) in an experimental model of cancer-related pain in mice, tolerant or not to morphine. The model of cancer-induced pain was used to evaluate these effects, with the injection of B16F10 cells, followed by the administration of the r-Phα1β, and evaluation of the mechanical threshold by the von Frey test. Also, adverse effects were assessed using a score scale, the rotarod, and open field tests. Results indicate that the administration of r-Phα1β provoked antinociception in animals with cancer-induced mechanical hyperalgesia, with or without morphine tolerance. Previous administration of r-Phα1β was able to recover the analgesic activity of morphine in animals tolerant to this opioid. r-Phα1β was proved safe for these parameters, as no adverse effects related to motor and behavioral activity were observed following intravenous administration. This study suggests that the concomitant use of morphine and r-Phα1β could be a viable strategy for pain modulation in cancer patients.
Collapse
Affiliation(s)
- Mariana de Melo Cardoso
- Laboratory of Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of the Extreme South Catarinense (UNESC), Criciúma, SC, Brazil; Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100, MG, Brazil
| | - Rahisa Scussel
- Laboratory of Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of the Extreme South Catarinense (UNESC), Criciúma, SC, Brazil
| | - Jéssica da Silva Abel
- Laboratory of Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of the Extreme South Catarinense (UNESC), Criciúma, SC, Brazil
| | - Fernando Oriques Pereira
- Laboratory of Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of the Extreme South Catarinense (UNESC), Criciúma, SC, Brazil
| | - Lidiane Anastácio Cruz
- Laboratory of Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of the Extreme South Catarinense (UNESC), Criciúma, SC, Brazil
| | - Franciane da Costa Constante
- Laboratory of Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of the Extreme South Catarinense (UNESC), Criciúma, SC, Brazil
| | - Ellen De Pieri
- Laboratory of Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of the Extreme South Catarinense (UNESC), Criciúma, SC, Brazil
| | - Helena Mendes Abelaira
- Laboratory of Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of the Extreme South Catarinense (UNESC), Criciúma, SC, Brazil
| | - Juliano Ferreira
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Marcus Vinícius Gomez
- Graduate Program in Health Sciences, Institute of Education and Research, Santa Casa de Belo Horizonte, Belo Horizonte, MG, Brazil; Center of Technology in Molecular Medicine, School of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Flávia Karine Rigo
- Laboratory of Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of the Extreme South Catarinense (UNESC), Criciúma, SC, Brazil
| | - Ricardo Andrez Machado-de-Ávila
- Laboratory of Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of the Extreme South Catarinense (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
3
|
Taylor NE, Ferrari L. Discovering chronic pain treatments: better animal models might help us get there. J Clin Invest 2023; 133:167814. [PMID: 36856117 PMCID: PMC9974092 DOI: 10.1172/jci167814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Abstract
Only three classes of pain medications have made it into clinical use in the past 60 years despite intensive efforts and the need for nonaddictive pain treatments. One reason for the failure involves the use of animal models that lack mechanistic similarity to human pain conditions, with endpoint measurements that may not reflect the human pain experience. In this issue of the JCI, Ding, Fischer, and co-authors developed the foramen lacerum impingement of trigeminal nerve root (FLIT) model of human trigeminal neuralgia that has improved face, construct, and predictive validities over those of current models. They used the FLIT model to investigate the role that abnormal, hypersynchronous cortical activity contributed to a neuropathic pain state. Unrestrained, synchronous glutamatergic activity in the primary somatosensory cortex upper lip and jaw (S1ULp-S1J) region of the somatosensory cortex drove pain phenotypes. The model establishes a powerful tool to continue investigating the interaction between the peripheral and central nervous systems that leads to chronic pain.
Collapse
Affiliation(s)
- Norman E. Taylor
- Department of Anesthesiology, The University of Utah, Salt Lake City, Utah, USA
| | - Luiz Ferrari
- Department of Anesthesiology, The University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
4
|
Eisenach JC, Rice AS. Improving Preclinical Development of Novel Interventions to Treat Pain: Insanity Is Doing the Same Thing Over and Over and Expecting Different Results. Anesth Analg 2022; 135:1128-1136. [PMID: 36384008 PMCID: PMC9976707 DOI: 10.1213/ane.0000000000006249] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Preclinical pain research has applied state-of-the-art methods over the past 40 years to describe, characterize, and image molecules, cells, and circuits in rodents to understand the pathophysiology of chronic pain. Despite generating a plethora of novel analgesic targets, pharmaceuticals for chronic pain treatment remain largely limited to the same 6 drug classes as present 40 years ago. It is possible that 40 years of effort has brought us to the verge of a paradigm shift and an explosion of novel analgesic drug classes with remarkable safety, efficacy, and tolerability. We think it more likely that advances will not occur until we follow the description of exciting discoveries with hypothesis testing using clinically relevant preclinical animal models and ethologically relevant outcome measures, which better reflect the clinical characteristics of chronic pain syndromes. Furthermore, to be valuable, experiments using such models must be conducted to the highest levels of internal validity, rigor, and reproducibility. Efforts by funders, most recently the Helping End Addiction Long-Term by the National Institutes of Health, aim to address some of these challenges and enhance communication and collaboration between preclinical and clinical investigators. However, the greater problem is a culture that emphasizes novelty and number of publications over scientific rigor and robust replication leading to a high likelihood of false-positive results. A path forward is provided by the evolution of clinical research beginning 50 years ago that resulted in methods to reduce bias and enhance transparency and ethics of reporting, moving from case reports to randomized controlled trials to innovative study designs with a focus on rigor, generalizability, and reproducibility. We argue that culture changed in clinical science in part because powerful forces outside the peer review system, especially from federal regulators that approve new drugs and human studies committees that addressed ethical failures of earlier research, mandated change in studies within their purview. Whether an external force will affect change in peclinical pain research is unclear.
Collapse
Affiliation(s)
- James C. Eisenach
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Andrew S.C. Rice
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, UK
| |
Collapse
|
5
|
Hagedorn JM, Engle AM, George TK, Karri J, Abdullah N, Ovrom E, Bocanegra-Becerra JE, D'Souza RS. An overview of painful diabetic peripheral neuropathy: Diagnosis and treatment advancements. Diabetes Res Clin Pract 2022; 188:109928. [PMID: 35580704 DOI: 10.1016/j.diabres.2022.109928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/12/2022] [Accepted: 05/09/2022] [Indexed: 01/09/2023]
Abstract
Diabetes mellitus remains a public health problem, affecting 422 million people worldwide. Currently, there is no consensus around treating painful diabetic peripheral neuropathy in a step-wise manner. Among the non-pharmacological interventions, neuromodulation has become a promising alternative. Over the past decade, significant clinical trials have paved the way for prompt inclusion of high-frequency spinal cord stimulation within the painful diabetic peripheral neuropathy treatment algorithm. This article aims to provide an updated evidence-based approach for the management of painful diabetic peripheral neuropathy.
Collapse
Affiliation(s)
| | - Alyson M Engle
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Jay Karri
- Johns Hopkins University, Baltimore, MD, USA
| | - Newaj Abdullah
- Division of Pain Medicine, Department of Anesthesiology, University of Utah, Salt Lake City, UT, USA
| | - Erik Ovrom
- Mayo Clinic Alix School of Medicine, Rochester, MN, USA
| | | | - Ryan S D'Souza
- Department of Anesthesiology and Perioperative Medicine, Division of Pain Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
6
|
Garcia-Caballero A, Gadotti VM, Ali MY, Bladen C, Gambeta E, Van Humbeck JF, MacCallum JL, Zamponi GW. A Synthetically Accessible Small-Molecule Inhibitor of USP5-Cav3.2 Calcium Channel Interactions with Analgesic Properties. ACS Chem Neurosci 2022; 13:524-536. [PMID: 35113527 DOI: 10.1021/acschemneuro.1c00765] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cav3.2 calcium channels are important mediators of nociceptive signaling in the primary afferent pain pathway, and their expression is increased in various rodent models of chronic pain. Previous work from our laboratory has shown that this is in part mediated by an aberrant expression of deubiquitinase USP5, which associates with these channels and increases their stability. Here, we report on a novel bioactive rhodanine compound (II-1), which was identified in compound library screens. II-1 inhibits biochemical interactions between USP5 and the Cav3.2 domain III-IV linker in a dose-dependent manner, without affecting the enzymatic activity of USP5. Molecular docking analysis reveals two potential binding pockets at the USP5-Cav3.2 interface that are distinct from the binding site of the deubiquitinase inhibitor WP1130 (a.k.a. degrasyn). With an understanding of the ability of some rhodanines to produce false positives in high-throughput screening, we have conducted several orthogonal assays to confirm the validity of this hit, including in vivo experiments. Intrathecal delivery of II-1 inhibited both phases of formalin-induced nocifensive behaviors in mice, as well as abolished thermal hyperalgesia induced by the delivery of complete Freund's adjuvant (CFA) to the hind paw. The latter effects were abolished in Cav3.2 null mice, thus confirming that Cav3.2 is required for the action of II-1. II-1 also mediated a robust inhibition of mechanical allodynia induced by injury to the sciatic nerve. Altogether, our data uncover a novel class of analgesics─well suited to rapid structure-activity relationship studies─that target the Cav3.2/USP5 interface.
Collapse
Affiliation(s)
- Agustin Garcia-Caballero
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary T2N 4N1, Canada
- Zymedyne Therapeutics, Calgary T2L 1Y8, Canada
| | - Vinicius M. Gadotti
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary T2N 4N1, Canada
- Zymedyne Therapeutics, Calgary T2L 1Y8, Canada
| | - Md Yousof Ali
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary T2N 4N1, Canada
| | - Chris Bladen
- Zymedyne Therapeutics, Calgary T2L 1Y8, Canada
- Faculty of Medicine, Macquarie University, 75 Talavera Rd, Sydney, New South Wales 2109, Australia
| | - Eder Gambeta
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary T2N 4N1, Canada
| | | | | | - Gerald W. Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary T2N 4N1, Canada
- Zymedyne Therapeutics, Calgary T2L 1Y8, Canada
| |
Collapse
|
7
|
Lanzetti S, Di Biase V. Small Molecules as Modulators of Voltage-Gated Calcium Channels in Neurological Disorders: State of the Art and Perspectives. Molecules 2022; 27:1312. [PMID: 35209100 PMCID: PMC8879281 DOI: 10.3390/molecules27041312] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 01/03/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) are widely expressed in the brain, heart and vessels, smooth and skeletal muscle, as well as in endocrine cells. VGCCs mediate gene transcription, synaptic and neuronal structural plasticity, muscle contraction, the release of hormones and neurotransmitters, and membrane excitability. Therefore, it is not surprising that VGCC dysfunction results in severe pathologies, such as cardiovascular conditions, neurological and psychiatric disorders, altered glycemic levels, and abnormal smooth muscle tone. The latest research findings and clinical evidence increasingly show the critical role played by VGCCs in autism spectrum disorders, Parkinson's disease, drug addiction, pain, and epilepsy. These findings outline the importance of developing selective calcium channel inhibitors and modulators to treat such prevailing conditions of the central nervous system. Several small molecules inhibiting calcium channels are currently used in clinical practice to successfully treat pain and cardiovascular conditions. However, the limited palette of molecules available and the emerging extent of VGCC pathophysiology require the development of additional drugs targeting these channels. Here, we provide an overview of the role of calcium channels in neurological disorders and discuss possible strategies to generate novel therapeutics.
Collapse
Affiliation(s)
| | - Valentina Di Biase
- Institute of Pharmacology, Department of Medical Statistics, Informatics and Health Economics, Medical University of Innsbruck, Peter-Mayr Strasse 1, A-6020 Innsbruck, Austria;
| |
Collapse
|
8
|
Gupta AK, Mena S, Jin Z, Gan TJ, Bergese S. Postoperative pain: a review of emerging therapeutic options. Expert Rev Neurother 2021; 21:1085-1100. [PMID: 34461794 DOI: 10.1080/14737175.2021.1974840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Postoperative pain is often managed by opioid medications, even though they carry a risk of adverse effects such as vomiting, constipation, sedation, respiratory depression and physical dependence. Furthermore, opioid use in the healthcare setting has likely contributed to the epidemic. However, the mismanagement of postoperative pain can result in delayed recovery time, impaired physical function, increased risk of morbidity and mortality, chronic pain, and higher healthcare costs. AREAS COVERED This review explores emerging therapeutic options and strategies in the management of acute postoperative pain and focuses on opioid-sparing, multimodal analgesia. This includes regional anesthetic techniques, non opioid pharmacotherapy, novel opioids and non-pharmacologic therapy. We have also discussed examples of novel analgesics and formulations which have potential benefits in reducing postoperative pain and opioid use after surgery. EXPERT OPINION The development of novel regional anesthesia techniques allows for opioid minimization in increasing number of surgical procedures. This synergizes with the availability of novel non-opioid analgesic adjucts. In addition, several novel opioid drugs have been developed which may be pathway selective and associated with less adverse effect than conventional opioids.
Collapse
Affiliation(s)
- Abhishek K Gupta
- Department of Anesthesiology, Stony Brook University Health Science Center, Stony Brook, United States
| | - Shayla Mena
- Department of Anesthesiology, Stony Brook University Health Science Center, Stony Brook, United States
| | - Zhaosheng Jin
- Department of Anesthesiology, Stony Brook University Health Science Center, Stony Brook, United States
| | - Tong J Gan
- Department of Anesthesiology, Stony Brook University Health Science Center, Stony Brook, United States
| | - Sergio Bergese
- Department of Anesthesiology, Stony Brook University Health Science Center, Stony Brook, United States.,Department of Neurological Surgery, Stony Brook University Health Science Center, Stony Brook, United States
| |
Collapse
|
9
|
Borges JP, Mekhail K, Fairn GD, Antonescu CN, Steinberg BE. Modulation of Pathological Pain by Epidermal Growth Factor Receptor. Front Pharmacol 2021; 12:642820. [PMID: 34054523 PMCID: PMC8149758 DOI: 10.3389/fphar.2021.642820] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/26/2021] [Indexed: 12/18/2022] Open
Abstract
Chronic pain has been widely recognized as a major public health problem that impacts multiple aspects of patient quality of life. Unfortunately, chronic pain is often resistant to conventional analgesics, which are further limited by their various side effects. New therapeutic strategies and targets are needed to better serve the millions of people suffering from this devastating disease. To this end, recent clinical and preclinical studies have implicated the epidermal growth factor receptor signaling pathway in chronic pain states. EGFR is one of four members of the ErbB family of receptor tyrosine kinases that have key roles in development and the progression of many cancers. EGFR functions by activating many intracellular signaling pathways following binding of various ligands to the receptor. Several of these signaling pathways, such as phosphatidylinositol 3-kinase, are known mediators of pain. EGFR inhibitors are known for their use as cancer therapeutics but given recent evidence in pilot clinical and preclinical investigations, may have clinical use for treating chronic pain. Here, we review the clinical and preclinical evidence implicating EGFR in pathological pain states and provide an overview of EGFR signaling highlighting how EGFR and its ligands drive pain hypersensitivity and interact with important pain pathways such as the opioid system.
Collapse
Affiliation(s)
- Jazlyn P Borges
- Neurosciences and Mental Health Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Katrina Mekhail
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Gregory D Fairn
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Costin N Antonescu
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Benjamin E Steinberg
- Neurosciences and Mental Health Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
10
|
Chalil A, Staudt MD, Harland TA, Leimer EM, Bhullar R, Argoff CE. A safety review of approved intrathecal analgesics for chronic pain management. Expert Opin Drug Saf 2021; 20:439-451. [PMID: 33583318 DOI: 10.1080/14740338.2021.1889513] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Introduction: Intrathecal (IT) drug therapy is an effective treatment option for patients with chronic pain of malignant or nonmalignant origin, with an established safety profile and fewer adverse effects compared to oral or parenteral pain medications. Morphine (a μ-opioid receptor agonist) and ziconotide (a non-opioid calcium channel antagonist) are the only IT agents approved by the U.S. Food and Drug Administration for the treatment of chronic pain. Although both are considered first-line IT therapies, each drug has unique properties and considerations.Areas Covered: This review will evaluate the pivotal trials that established the use of morphine and ziconotide as first-line IT therapy for patients with chronic pain, as well as safety and efficacy data generated from various retrospective and prospective studies.Expert Opinion: Morphine and ziconotide are effective IT therapies for patients with chronic malignant or nonmalignant pain that is refractory to other interventions. IT ziconotide is recommended as a first-line therapy due to its efficacy and avoidance of many adverse effects commonly associated with opioids. The use of IT morphine is also considered first-line; however, the risks of respiratory depression, withdrawal with drug discontinuation or pump malfunction, and the development of tolerance require careful patient selection and management.
Collapse
Affiliation(s)
- Alan Chalil
- Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, Ontario, Canada
| | - Michael D Staudt
- Department of Neurosurgery, Oakland University William Beaumont School of Medicine, Rochester, Michigan, USA.,Michigan Head and Spine Institute, Southfield, Michigan, USA
| | - Tessa A Harland
- Department of Neurosurgery, Albany Medical College, Albany, New York, USA
| | - Elizabeth M Leimer
- Department of Anesthesiology & Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Ravneet Bhullar
- Department of Anesthesiology, Albany Medical College, Albany, New York, USA
| | - Charles E Argoff
- Department of Neurology, Albany Medical College, Albany, New York
| |
Collapse
|
11
|
Banik RK, Engle MP. Ziconotide for Management of Cancer Pain Refractory to Pharmacotherapy: An Update. PAIN MEDICINE 2020; 21:3253-3259. [PMID: 32940675 DOI: 10.1093/pm/pnaa251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Ratan K Banik
- Department of Anesthesiology, University of Minnesota, Minneapolis, Minnesota
| | - Mitchell P Engle
- Institute of Precision Pain Medicine, Corpus Christi, Texas, USA
| |
Collapse
|
12
|
Chakraborty K, Joy M. High-value compounds from the molluscs of marine and estuarine ecosystems as prospective functional food ingredients: An overview. Food Res Int 2020; 137:109637. [PMID: 33233216 PMCID: PMC7457972 DOI: 10.1016/j.foodres.2020.109637] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/02/2020] [Accepted: 08/21/2020] [Indexed: 12/20/2022]
Abstract
Extensive biodiversity and availability of marine and estuarine molluscs, along with their their wide-range of utilities as food and nutraceutical resources developed keen attention of the food technologists and dieticians, particularly during the recent years. The current review comprehensively summarized the nutritional qualities, functional food attributes, and bioactive properties of these organisms. Among the phylum mollusca, Cephalopoda, Bivalvia, and Gastropoda were mostly reported for their nutraceutical applications and bioactive properties. The online search tools, like Scifinder/Science Direct/PubMed/Google Scholar/MarinLit database and marine natural product reports (1984-2019) were used to comprehend the information about the molluscs. More than 1334 secondary metabolites were reported from marine molluscs between the periods from 1984 to 2019. Among various classes of specialized metabolites, terpenes were occupied by 55% in gastropods, whereas sterols occupied 41% in bivalves. The marketed nutraceuticals, such as CadalminTM green mussel extract (Perna viridis) and Lyprinol® (Perna canaliculus) were endowed with potential anti-inflammatory activities, and were used against arthritis. Molluscan-derived therapeutics, for example, ziconotide was used as an analgesic, and elisidepsin was used in the treatment of cancer. Greater numbers of granted patents (30%) during 2016-2019 recognized the increasing importance of bioactive compounds from molluscs. Consumption of molluscs as daily diets could be helpful in the enhancement of immunity, and reduce the risk of several ailments. The present review comprehended the high value compounds and functional food ingredients from marine and estuarine molluscs.
Collapse
Affiliation(s)
- Kajal Chakraborty
- Marine Biotechnology Division, Central Marine Fisheries Research Institute, Ernakulam North, P.B. No. 1603, Cochin-682018, Kerala, India.
| | - Minju Joy
- Marine Biotechnology Division, Central Marine Fisheries Research Institute, Ernakulam North, P.B. No. 1603, Cochin-682018, Kerala, India
| |
Collapse
|
13
|
McDowell GC, Saulino MF, Wallace M, Grigsby EJ, Rauck RL, Kim P, Vanhove GF, Ryan R, Huang IZ, Deer T. Effectiveness and Safety of Intrathecal Ziconotide: Final Results of the Patient Registry of Intrathecal Ziconotide Management (PRIZM). PAIN MEDICINE 2020; 21:2925-2938. [DOI: 10.1093/pm/pnaa115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Abstract
Background and Objectives
The Patient Registry of Intrathecal Ziconotide Management evaluated the long-term effectiveness and safety of intrathecal ziconotide.
Methods
The study was a prospective, multicenter observational study of intrathecal ziconotide in US clinical practice. Patients were adults with severe chronic pain that warranted intrathecal therapy. Ziconotide was initiated as the single agent in the pump; however, other intrathecal medications were permitted. The primary efficacy outcome was ≥30% reduction in numeric pain rating scale score from baseline at week 12. A secondary outcome was patient global impression of change. Adverse events were solicited at each visit.
Results
The registry enrolled 93 patients. Seventy-four and 28 patients completed 12 weeks and 18 months of treatment, respectively. In the overall patient population, 17.4% had ≥30% pain reduction from baseline at week 12, with a mean reduction in pain of 10.9%. At month 18, 38.5% of patients had ≥30% pain reduction from baseline, with a mean pain reduction of 24.7%. Patient-rated improvement was reported in 67% of patients at week 12 and 71% at month 18. Almost all patients experienced adverse events, the most common of which were nausea (25.8%), confusional state (22.6%), and dizziness (20.4%).
Conclusions
Final study analyses showed that intrathecal ziconotide provided clinically meaningful pain relief in 17.4% and 38.5% of patients at week 12 and month 18, respectively. At these same time points, patient-rated improvement was reported in at least two-thirds of patients. The safety profile was consistent with that listed in the ziconotide prescribing information.
Collapse
Affiliation(s)
| | | | - Mark Wallace
- University of California, San Diego, La Jolla, California
| | | | - Richard L Rauck
- Carolinas Pain Institute, The Center for Clinical Research, Winston-Salem, North Carolina
| | - Philip Kim
- Center for Interventional Pain and Spine, LLC, Newark, Delaware
| | - Geertrui F Vanhove
- ‖
- Surrozen Inc, South San Francisco, California; **Jazz Pharmaceuticals, Palo Alto, California; ††Grail Inc, Menlo Park, California
| | - Robert Ryan
- ‖
- Surrozen Inc, South San Francisco, California; **Jazz Pharmaceuticals, Palo Alto, California; ††Grail Inc, Menlo Park, California
| | - I-Zu Huang
- ‖
- Surrozen Inc, South San Francisco, California; **Jazz Pharmaceuticals, Palo Alto, California; ††Grail Inc, Menlo Park, California
| | - Timothy Deer
- The Spine and Nerve Center of the Virginias, Charleston, West Virginia, USA
| |
Collapse
|
14
|
Rigo FK, Rossato MF, Borges V, da Silva JF, Pereira EMR, de Ávila RAM, Trevisan G, Dos Santos DC, Diniz DM, Silva MAR, de Castro CJ, Cunha TM, Ferreira J, Gomez MV. Analgesic and side effects of intravenous recombinant Phα1β. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20190070. [PMID: 32362927 PMCID: PMC7179342 DOI: 10.1590/1678-9199-jvatitd-2019-0070] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Background Intrathecal injection of voltage-sensitive calcium channel blocker peptide toxins exerts analgesic effect in several animal models of pain. Upon intrathecal administration, recombinant Phα1β exerts the same analgesic effects as the those of the native toxin. However, from a clinical perspective, the intrathecal administration limits the use of anesthetic drugs in patients. Therefore, this study aimed to investigate the possible antinociceptive effect of intravenous recombinant Phα1β in rat models of neuropathic pain, as well as its side effects on motor, cardiac (heart rate and blood pressure), and biochemical parameters. Methods Male Wistar rats and male Balb-C mice were used in this study. Giotto Biotech® synthesized the recombinant version of Phα1β using Escherichia coli expression. In rats, neuropathic pain was induced by chronic constriction of the sciatic nerve and paclitaxel-induced acute and chronic pain. Mechanical sensitivity was evaluated using von Frey filaments. A radiotelemeter transmitter (TA11PA-C10; Data Sciences, St. Paul, MN, USA) was placed on the left carotid of mice for investigation of cardiovascular side effects. Locomotor activity data were evaluated using the open-field paradigm, and serum CKMB, TGO, TGP, LDH, lactate, creatinine, and urea levels were examined. Results Intravenous administration of recombinant Phα1β toxin induced analgesia for up to 4 h, with ED50 of 0.02 (0.01-0.03) mg/kg, and reached the maximal effect (Emax = 100% antinociception) at a dose of 0.2 mg/kg. No significant changes were observed in any of the evaluated motor, cardiac or biochemical parameters. Conclusion Our data suggest that intravenous administration of recombinant Phα1β may be feasible for drug-induced analgesia, without causing any severe side effects.
Collapse
Affiliation(s)
- Flavia Karine Rigo
- Graduate Program in Health Sciences, University of the Extreme South of Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Mateus Fortes Rossato
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Vanessa Borges
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Juliana Figueira da Silva
- Institute of Education and Research of Santa Casa Belo Horizonte, Santa Casa of Belo Horizonte Group, Belo Horizonte, MG, Brazil
| | - Elizete Maria Rita Pereira
- Institute of Education and Research of Santa Casa Belo Horizonte, Santa Casa of Belo Horizonte Group, Belo Horizonte, MG, Brazil
| | | | - Gabriela Trevisan
- Graduate Program in Health Sciences, University of the Extreme South of Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Duana Carvalho Dos Santos
- Institute of Education and Research of Santa Casa Belo Horizonte, Santa Casa of Belo Horizonte Group, Belo Horizonte, MG, Brazil
| | - Danuza Montijo Diniz
- Institute of Education and Research of Santa Casa Belo Horizonte, Santa Casa of Belo Horizonte Group, Belo Horizonte, MG, Brazil
| | - Marco Aurélio Romano Silva
- Department of Neurosciences, School of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Célio José de Castro
- Institute of Education and Research of Santa Casa Belo Horizonte, Santa Casa of Belo Horizonte Group, Belo Horizonte, MG, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Juliano Ferreira
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Marcus Vinicius Gomez
- Institute of Education and Research of Santa Casa Belo Horizonte, Santa Casa of Belo Horizonte Group, Belo Horizonte, MG, Brazil
| |
Collapse
|
15
|
Brinzeu A, Berthiller J, Caillet J, Staquet H, Mertens P. Ziconotide for spinal cord injury‐related pain. Eur J Pain 2019; 23:1688-1700. [DOI: 10.1002/ejp.1445] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 05/27/2019] [Accepted: 06/20/2019] [Indexed: 01/18/2023]
Affiliation(s)
- Andrei Brinzeu
- Neurosurgical Department Pierre Wertheimer Hospital, Hospices Civils de Lyon, Lyon 1 University Lyon France
- Pain Center “Pierre Wertheimer” Hospital, Hospices Civils de Lyon Lyon France
- University of Medicine and Pharmacy “Victor Babes” Timisoara Timisoara Romania
| | - Julien Berthiller
- Neurosurgical Department Pierre Wertheimer Hospital, Hospices Civils de Lyon, Lyon 1 University Lyon France
| | | | - Helene Staquet
- Neurosurgical Department Pierre Wertheimer Hospital, Hospices Civils de Lyon, Lyon 1 University Lyon France
- Neurosurgical Department Beaujon University Hospital‐APHP Clichy France
| | - Patrick Mertens
- Neurosurgical Department Pierre Wertheimer Hospital, Hospices Civils de Lyon, Lyon 1 University Lyon France
- Pain Center “Pierre Wertheimer” Hospital, Hospices Civils de Lyon Lyon France
| |
Collapse
|
16
|
Marine Toxins and Nociception: Potential Therapeutic Use in the Treatment of Visceral Pain Associated with Gastrointestinal Disorders. Toxins (Basel) 2019; 11:toxins11080449. [PMID: 31370176 PMCID: PMC6723473 DOI: 10.3390/toxins11080449] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022] Open
Abstract
Visceral pain, of which the pathogenic basis is currently largely unknown, is a hallmark symptom of both functional disorders, such as irritable bowel syndrome, and inflammatory bowel disease. Intrinsic sensory neurons in the enteric nervous system and afferent sensory neurons of the dorsal root ganglia, connecting with the central nervous system, represent the primary neuronal pathways transducing gut visceral pain. Current pharmacological therapies have several limitations, owing to their partial efficacy and the generation of severe adverse effects. Numerous cellular targets of visceral nociception have been recognized, including, among others, channels (i.e., voltage-gated sodium channels, VGSCs, voltage-gated calcium channels, VGCCs, Transient Receptor Potential, TRP, and Acid-sensing ion channels, ASICs) and neurotransmitter pathways (i.e., GABAergic pathways), which represent attractive targets for the discovery of novel drugs. Natural biologically active compounds, such as marine toxins, able to bind with high affinity and selectivity to different visceral pain molecular mediators, may represent a useful tool (1) to improve our knowledge of the physiological and pathological relevance of each nociceptive target, and (2) to discover therapeutically valuable molecules. In this review we report the most recent literature describing the effects of marine toxin on gastrointestinal visceral pain pathways and the possible clinical implications in the treatment of chronic pain associated with gut diseases.
Collapse
|
17
|
Safavi-Hemami H, Brogan SE, Olivera BM. Pain therapeutics from cone snail venoms: From Ziconotide to novel non-opioid pathways. J Proteomics 2018; 190:12-20. [PMID: 29777871 DOI: 10.1016/j.jprot.2018.05.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/15/2018] [Indexed: 01/04/2023]
Abstract
There have been numerous attempts to develop non-opioid drugs for severe pain, but the vast majority of these efforts have failed. A notable exception is Ziconotide (Prialt®), approved by the FDA in 2004. In this review, we summarize the present status of Ziconotide as a therapeutic drug and introduce a wider framework: the potential of venom peptides from cone snails as a resource providing a continuous pipeline for the discovery of non-opioid pain therapeutics. An auxiliary theme that we hope to develop is that these venoms, already a validated starting point for non-opioid drug leads, should also provide an opportunity for identifying novel molecular targets for future pain drugs. This review comprises several sections: the first focuses on Ziconotide as a therapeutic (including a historical retrospective and a clinical perspective); followed by sections on other promising Conus venom peptides that are either in clinical or pre-clinical development. We conclude with a discussion on why the outlook for discovery appears exceptionally promising. The combination of new technologies in diverse fields, including the development of novel high-content assays and revolutionary advancements in transcriptomics and proteomics, puts us at the cusp of providing a continuous pipeline of non-opioid drug innovations for pain. SIGNIFICANCE: The current opioid epidemic is the deadliest drug crisis in American history. Thus, this review on the discovery of non-opioid pain therapeutics and pathways from cone snail venoms is significant and timely.
Collapse
Affiliation(s)
| | - Shane E Brogan
- Anesthesiology, University of Utah, Salt Lake City, UT, United States; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Baldomero M Olivera
- Departments of Biology, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
18
|
Abstract
Neuropathic pain represents a significant and mounting burden on patients and society at large. Management of neuropathic pain, however, is both intricate and challenging, exacerbated by the limited quantity and quality of clinically available treatments. On this stage, dysfunctional voltage-gated ion channels, especially the presynaptic N-type voltage-gated calcium channel (VGCC) (Cav2.2) and the tetrodotoxin-sensitive voltage-gated sodium channel (VGSC) (Nav1.7), underlie the pathophysiology of neuropathic pain and serve as high profile therapeutic targets. Indirect regulation of these channels holds promise for the treatment of neuropathic pain. In this review, we focus on collapsin response mediator protein 2 (CRMP2), a protein with emergent roles in voltage-gated ion channel trafficking and discuss the therapeutic potential of targetting this protein.
Collapse
|
19
|
Racemic X-ray structure of L-type calcium channel antagonist Calciseptine prepared by total chemical synthesis. Sci China Chem 2018. [DOI: 10.1007/s11426-017-9198-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
20
|
Maatoug R, Jebali J, Guieu R, De Waard M, Kharrat R. BotAF, a new Buthus occitanus tunetanus scorpion toxin, produces potent analgesia in rodents. Toxicon 2018; 149:72-85. [PMID: 29337220 DOI: 10.1016/j.toxicon.2018.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 11/25/2022]
Abstract
This work reports the purification of new potent scorpion neuropeptide, named BotAF, by an activity-guided screening approach. BotAF is a 64-residue long-chain peptide that shares very high similarity with the original β-like scorpion toxin group, in which several peptides have been characterized to be anti-nociceptive in rodents. BotAF administration to rodents does not produce any toxicity or motor impairment, including at high doses. In all models investigated, BotAF turned out to be an efficient peptide in abolishing acute and inflammatory (both somatic and visceral) pain in rodents. It performs with high potency compared to standard analgesics tested in the same conditions. The anti-nociceptive activity of BotAF depends on the route of injection: it is inactive when tested by i.c.v. or i.v. routes but gains in potency when pre-injected locally (in the same compartment than the irritant itself) or by i.t. root 40 to 60 min before pain induction, respectively. BotAF is not an AINS-like compound as it fails to reduce inflammatory edema. Also, it does not activate the opioidergic system as its activity is not affected by naloxone. BotAF does also not bind onto RyR and has low activity towards DRG ion channels (particularly TTX sensitive Na+ channels) and does not bind onto rat brain synaptosome receptors. In somatic and visceral pain models, BotAF dose-dependently inhibited lumbar spinal cord c-fos/c-jun mRNA up regulation. Altogether, our data favor a spinal or peripheral anti-nociceptive mode of action of BotAF.
Collapse
Affiliation(s)
- Riadh Maatoug
- Université de Tunis El Manar, Institut Pasteur de Tunis, Laboratoire des Venins et Biomolécules Thérapeutiques, 13, Place Pasteur BP-74, Tunis, 1002, Tunisia
| | - Jed Jebali
- Université de Tunis El Manar, Institut Pasteur de Tunis, Laboratoire des Venins et Biomolécules Thérapeutiques, 13, Place Pasteur BP-74, Tunis, 1002, Tunisia
| | - Régis Guieu
- Biochimie, Hôpital de la Timone, 13005, Marseille, France
| | - Michel De Waard
- Inserm U1087, Institut du Thorax, groupe IIb, Université de Nantes, 8 quai moncousu, 44000, Nantes, France; Smartox Biotechnology, 570 rue de la chimie, bâtiment Nanobio, 38700, Saint Martin d'Hères, France
| | - Riadh Kharrat
- Université de Tunis El Manar, Institut Pasteur de Tunis, Laboratoire des Venins et Biomolécules Thérapeutiques, 13, Place Pasteur BP-74, Tunis, 1002, Tunisia.
| |
Collapse
|
21
|
SUMOylation and calcium signalling: potential roles in the brain and beyond. Neuronal Signal 2017; 1:NS20160010. [PMID: 32714579 PMCID: PMC7373246 DOI: 10.1042/ns20160010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 12/23/2022] Open
Abstract
Small ubiquitin-like modifier (SUMO) conjugation (or SUMOylation) is a post-translational protein modification implicated in alterations to protein expression, localization and function. Despite a number of nuclear roles for SUMO being well characterized, this process has only started to be explored in relation to membrane proteins, such as ion channels. Calcium ion (Ca2+) signalling is crucial for the normal functioning of cells and is also involved in the pathophysiological mechanisms underlying relevant neurological and cardiovascular diseases. Intracellular Ca2+ levels are tightly regulated; at rest, most Ca2+ is retained in organelles, such as the sarcoplasmic reticulum, or in the extracellular space, whereas depolarization triggers a series of events leading to Ca2+ entry, followed by extrusion and reuptake. The mechanisms that maintain Ca2+ homoeostasis are candidates for modulation at the post-translational level. Here, we review the effects of protein SUMOylation, including Ca2+ channels, their proteome and other proteins associated with Ca2+ signalling, on vital cellular functions, such as neurotransmission within the central nervous system (CNS) and in additional systems, most prominently here, in the cardiac system.
Collapse
|
22
|
Netirojjanakul C, Miranda LP. Progress and challenges in the optimization of toxin peptides for development as pain therapeutics. Curr Opin Chem Biol 2017; 38:70-79. [PMID: 28376346 DOI: 10.1016/j.cbpa.2017.03.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 02/25/2017] [Accepted: 03/13/2017] [Indexed: 02/05/2023]
Abstract
The number of new toxin peptide discoveries has been rapidly growing in the past few decades. Because of progress in proteomics, sequencing technologies, and high throughput bioassays, the search for new toxin peptides from venom collections and potency optimization has become manageable. However, to date, only six toxin peptide-derived therapeutics have been approved by the USFDA, with only one, ziconotide, for a pain indication. The challenge of venom-derived peptide therapeutic development remains in improving selectivity to the target and more importantly, in delivery of these peptides to the sites of action in the central and peripheral nervous system. In this review, we highlight peptide toxins that target major therapeutic targets for pain and discuss the challenges of developing toxin peptides as potential therapeutics.
Collapse
Affiliation(s)
- Chawita Netirojjanakul
- Therapeutic Discovery, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | - Les P Miranda
- Therapeutic Discovery, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| |
Collapse
|
23
|
Moutal A, Li W, Wang Y, Ju W, Luo S, Cai S, François-Moutal L, Perez-Miller S, Hu J, Dustrude ET, Vanderah TW, Gokhale V, Khanna M, Khanna R. Homology-guided mutational analysis reveals the functional requirements for antinociceptive specificity of collapsin response mediator protein 2-derived peptides. Br J Pharmacol 2017; 175:2244-2260. [PMID: 28161890 DOI: 10.1111/bph.13737] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/26/2017] [Accepted: 01/31/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE N-type voltage-gated calcium (Cav 2.2) channels are critical determinants of increased neuronal excitability and neurotransmission accompanying persistent neuropathic pain. Although Cav 2.2 channel antagonists are recommended as first-line treatment for neuropathic pain, calcium-current blocking gabapentinoids inadequately alleviate chronic pain symptoms and often exhibit numerous side effects. Collapsin response mediator protein 2 (CRMP2) targets Cav 2.2 channels to the sensory neuron membrane and allosterically modulates their function. A 15-amino-acid peptide (CBD3), derived from CRMP2, disrupts the functional protein-protein interaction between CRMP2 and Cav 2.2 channels to inhibit calcium influx, transmitter release and acute, inflammatory and neuropathic pain. Here, we have mapped the minimal domain of CBD3 necessary for its antinociceptive potential. EXPERIMENTAL APPROACH Truncated as well as homology-guided mutant versions of CBD3 were generated and assessed using depolarization-evoked calcium influx in rat dorsal root ganglion neurons, binding between CRMP2 and Cav 2.2 channels, whole-cell voltage clamp electrophysiology and behavioural effects in two models of experimental pain: post-surgical pain and HIV-induced sensory neuropathy induced by the viral glycoprotein 120. KEY RESULTS The first six amino acids within CBD3 accounted for all in vitro activity and antinociception. Spinal administration of a prototypical peptide (TAT-CBD3-L5M) reversed pain behaviours. Homology-guided mutational analyses of these six amino acids identified at least two residues, Ala1 and Arg4, as being critical for antinociception in two pain models. CONCLUSIONS AND IMPLICATIONS These results identify an antinociceptive scaffold core in CBD3 that can be used for development of low MW mimetics of CBD3. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
- Aubin Moutal
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Wennan Li
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Yue Wang
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Weina Ju
- Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China.,Department of Pharmacology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Shizhen Luo
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Song Cai
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | | | | | - Jackie Hu
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Erik T Dustrude
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Todd W Vanderah
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Vijay Gokhale
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - May Khanna
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Rajesh Khanna
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA.,Neuroscience Graduate Interdisciplinary Program, College of Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
24
|
(S)-lacosamide inhibition of CRMP2 phosphorylation reduces postoperative and neuropathic pain behaviors through distinct classes of sensory neurons identified by constellation pharmacology. Pain 2017; 157:1448-1463. [PMID: 26967696 DOI: 10.1097/j.pain.0000000000000555] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Chronic pain affects the life of millions of people. Current treatments have deleterious side effects. We have advanced a strategy for targeting protein interactions which regulate the N-type voltage-gated calcium (CaV2.2) channel as an alternative to direct channel block. Peptides uncoupling CaV2.2 interactions with the axonal collapsin response mediator protein 2 (CRMP2) were antinociceptive without effects on memory, depression, and reward/addiction. A search for small molecules that could recapitulate uncoupling of the CaV2.2-CRMP2 interaction identified (S)-lacosamide [(S)-LCM], the inactive enantiomer of the Food and Drug Administration-approved antiepileptic drug (R)-lacosamide [(R)-LCM, Vimpat]. We show that (S)-LCM, but not (R)-LCM, inhibits CRMP2 phosphorylation by cyclin dependent kinase 5, a step necessary for driving CaV2.2 activity, in sensory neurons. (S)-lacosamide inhibited depolarization-induced Ca influx with a low micromolar IC50. Voltage-clamp electrophysiology experiments demonstrated a commensurate reduction in Ca currents in sensory neurons after an acute application of (S)-LCM. Using constellation pharmacology, a recently described high content phenotypic screening platform for functional fingerprinting of neurons that uses subtype-selective pharmacological agents to elucidate cell-specific combinations (constellations) of key signaling proteins that define specific cell types, we investigated if (S)-LCM preferentially acts on certain types of neurons. (S)-lacosamide decreased the dorsal root ganglion neurons responding to mustard oil, and increased the number of cells responding to menthol. Finally, (S)-LCM reversed thermal hypersensitivity and mechanical allodynia in a model of postoperative pain, and 2 models of neuropathic pain. Thus, using (S)-LCM to inhibit CRMP2 phosphorylation is a novel and efficient strategy to treat pain, which works by targeting specific sensory neuron populations.
Collapse
|
25
|
Sustained relief of ongoing experimental neuropathic pain by a CRMP2 peptide aptamer with low abuse potential. Pain 2017; 157:2124-2140. [PMID: 27537210 DOI: 10.1097/j.pain.0000000000000628] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Uncoupling the protein-protein interaction between collapsin response mediator protein 2 (CRMP2) and N-type voltage-gated calcium channel (CaV2.2) with an allosteric CRMP2-derived peptide (CBD3) is antinociceptive in rodent models of inflammatory and neuropathic pain. We investigated the efficacy, duration of action, abuse potential, and neurobehavioral toxicity of an improved mutant CRMP2 peptide. A homopolyarginine (R9)-conjugated CBD3-A6K (R9-CBD3-A6K) peptide inhibited the CaV2.2-CRMP2 interaction in a concentration-dependent fashion and diminished surface expression of CaV2.2 and depolarization-evoked Ca influx in rat dorsal root ganglia neurons. In vitro studies demonstrated suppression of excitability of small-to-medium diameter dorsal root ganglion and inhibition of subtypes of voltage-gated Ca channels. Sprague-Dawley rats with tibial nerve injury had profound and long-lasting tactile allodynia and ongoing pain. Immediate administration of R9-CBD3-A6K produced enhanced dopamine release from the nucleus accumbens shell selectively in injured animals, consistent with relief of ongoing pain. R9-CBD3-A6K, when administered repeatedly into the central nervous system ventricles of naive rats, did not result in a positive conditioned place preference demonstrating a lack of abusive liability. Continuous subcutaneous infusion of R9-CBD3-A6K over a 24- to 72-hour period reversed tactile allodynia and ongoing pain, demonstrating a lack of tolerance over this time course. Importantly, continuous infusion of R9-CBD3-A6K did not affect motor activity, anxiety, depression, or memory and learning. Collectively, these results validate the potential therapeutic significance of targeting the CaV-CRMP2 axis for treatment of neuropathic pain.
Collapse
|
26
|
Massonnet P, Haler JRN, Upert G, Degueldre M, Morsa D, Smargiasso N, Mourier G, Gilles N, Quinton L, De Pauw E. Ion Mobility-Mass Spectrometry as a Tool for the Structural Characterization of Peptides Bearing Intramolecular Disulfide Bond(s). JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2016; 27:1637-1646. [PMID: 27488317 DOI: 10.1007/s13361-016-1443-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 06/19/2016] [Accepted: 06/24/2016] [Indexed: 06/06/2023]
Abstract
Disulfide bonds are post-translationnal modifications that can be crucial for the stability and the biological activities of natural peptides. Considering the importance of these disulfide bond-containing peptides, the development of new techniques in order to characterize these modifications is of great interest. For this purpose, collision cross cections (CCS) of a large data set of 118 peptides (displaying various sequences) bearing zero, one, two, or three disulfide bond(s) have been measured in this study at different charge states using ion mobility-mass spectrometry. From an experimental point of view, CCS differences (ΔCCS) between peptides bearing various numbers of disulfide bonds and peptides having no disulfide bonds have been calculated. The ΔCCS calculations have also been applied to peptides bearing two disulfide bonds but different cysteine connectivities (Cys1-Cys2/Cys3-Cys4; Cys1-Cys3/Cys2-Cys4; Cys1-Cys4/Cys2-Cys3). The effect of the replacement of a proton by a potassium adduct on a peptidic structure has also been investigated. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Philippe Massonnet
- Laboratory of Mass Spectrometry, University of Liege, Quartier Agora, Allée du six Aout 11, B-4000, Liege, Belgium
| | - Jean R N Haler
- Laboratory of Mass Spectrometry, University of Liege, Quartier Agora, Allée du six Aout 11, B-4000, Liege, Belgium
| | - Gregory Upert
- Commissariat à l'Energie Atomique, DRF/iBiTec-S/SIMOPRO, CE Saclay, 91191, Gif-sur-Yvette, France
| | - Michel Degueldre
- Laboratory of Mass Spectrometry, University of Liege, Quartier Agora, Allée du six Aout 11, B-4000, Liege, Belgium
| | - Denis Morsa
- Laboratory of Mass Spectrometry, University of Liege, Quartier Agora, Allée du six Aout 11, B-4000, Liege, Belgium
| | - Nicolas Smargiasso
- Laboratory of Mass Spectrometry, University of Liege, Quartier Agora, Allée du six Aout 11, B-4000, Liege, Belgium
| | - Gilles Mourier
- Commissariat à l'Energie Atomique, DRF/iBiTec-S/SIMOPRO, CE Saclay, 91191, Gif-sur-Yvette, France
| | - Nicolas Gilles
- Commissariat à l'Energie Atomique, DRF/iBiTec-S/SIMOPRO, CE Saclay, 91191, Gif-sur-Yvette, France
| | - Loïc Quinton
- Laboratory of Mass Spectrometry, University of Liege, Quartier Agora, Allée du six Aout 11, B-4000, Liege, Belgium
| | - Edwin De Pauw
- Laboratory of Mass Spectrometry, University of Liege, Quartier Agora, Allée du six Aout 11, B-4000, Liege, Belgium.
| |
Collapse
|
27
|
Prusik J, Argoff C, Peng S, Pilitsis JG. Use of Low Dose Ziconotide as First-Line Intrathecal Monotherapy. Neuromodulation 2016; 20:386-391. [DOI: 10.1111/ner.12486] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/12/2016] [Accepted: 07/12/2016] [Indexed: 02/01/2023]
Affiliation(s)
- Julia Prusik
- Department of Neurosurgery; Albany Medical Center; Albany NY USA
- Department of Neurology; Albany Medical Center; Albany NY USA
- Department of Neuroscience and Experimental Therapeutics; Albany Medical College; Albany NY USA
| | - Charles Argoff
- Department of Neurology; Albany Medical Center; Albany NY USA
| | - Sophia Peng
- Department of Neurosurgery; Albany Medical Center; Albany NY USA
| | - Julie G. Pilitsis
- Department of Neurosurgery; Albany Medical Center; Albany NY USA
- Department of Neuroscience and Experimental Therapeutics; Albany Medical College; Albany NY USA
| |
Collapse
|
28
|
Sivanesan E, Gitlin MC. Desmoid Tumors: A Review of the Literature and Pharmacologic Management. J Pain Palliat Care Pharmacother 2016; 30:99-105. [PMID: 27159399 DOI: 10.3109/15360288.2016.1167807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Desmoid tumors represent a nonmalignant proliferation of fibroblast-related cells. These rare tumors are difficult to treat and often persist as indolent, lifelong conditions. There are a number of treatments available for both anatomic and symptom regression. Some of these treatments, unfortunately, may not provide long-lasting results and may result in further complications. Pain is a distressing symptom that may be due to the tumor itself or the result of utilized treatments. Pharmacologic therapies represent a noninvasive alternative to surgical resection. Pain symptoms require therapeutic regimens that must be modified as the tumor evolves in expression. The individualized pain treatment program utilized may often reflect principles used in both nonmalignant and malignant pain management models. This review seeks to increase awareness of desmoid tumors through a review of the literature and discussion of its pharmacotherapeutic management.
Collapse
|
29
|
Pope JE, Deer TR, Bruel BM, Falowski S. Clinical Uses of Intrathecal Therapy and Its Placement in the Pain Care Algorithm. Pain Pract 2016; 16:1092-1106. [PMID: 26914961 DOI: 10.1111/papr.12438] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 11/17/2015] [Accepted: 12/02/2015] [Indexed: 11/29/2022]
Abstract
Intrathecal drug delivery is an effective treatment option for patients with severe chronic pain who have not obtained adequate analgesia from more conservative therapies (eg, physical therapy, systemic opioids, nonsteroidal anti-inflammatory drugs, antidepressants, and anticonvulsants). This review focuses on, but is not limited to, the 2 agents currently approved by the U.S. Food and Drug Administration for intrathecal analgesia: preservative-free morphine and ziconotide (a nonopioid, selective N-type calcium channel blocker). We describe the appropriate use of intrathecal therapy in the management of severe chronic pain, based on current best practices. Topics addressed here include patient selection, trialing, dosing and titration, adverse event profiles, long-term management, intrathecal therapy for cancer-related pain, and the placement of intrathecal therapy in the pain care algorithm. In appropriately selected patients with chronic pain, intrathecal therapy can provide substantial pain relief with improved functioning and quality of life. Successful long-term management requires ongoing patient monitoring for changes in efficacy and the occurrence of adverse events, with subsequent changes in intrathecal dosing and titration, the addition of adjuvant intrathecal agents, and the use of concomitant oral medications to address side effects, as needed. Based on an infrequent but clinically concerning risk of overdose, granuloma, and other opioid-induced complications, nonopioid therapy with ziconotide may be preferred as a first-line intrathecal therapy in patients without a history of psychosis or allergy.
Collapse
Affiliation(s)
- Jason E Pope
- Summit Pain Alliance, Santa Rosa, California, U.S.A
| | - Timothy R Deer
- Center for Pain Relief, Charleston, West Virginia, U.S.A
| | - Brian M Bruel
- University of Texas, M.D. Anderson Cancer Center, Houston, Texas, U.S.A
| | - Steven Falowski
- St. Luke's Neurosurgical Associates, St. Luke's University Health Network, Bethlehem, Pennsylvania, U.S.A
| |
Collapse
|
30
|
McDowell GC, Pope JE. Intrathecal Ziconotide: Dosing and Administration Strategies in Patients With Refractory Chronic Pain. Neuromodulation 2016; 19:522-32. [PMID: 26856969 PMCID: PMC5067570 DOI: 10.1111/ner.12392] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/10/2015] [Indexed: 11/30/2022]
Abstract
Introduction Ziconotide is a non‐opioid analgesic for intrathecal (IT) administration. The aim of this review is to provide a comprehensive and clinically relevant summary of the literature on dosing and administration with IT ziconotide in the management of refractory chronic pain, and to describe novel dosing strategies intended to improve clinical outcomes. Materials and Methods A Medline search was conducted for “ziconotide,” supplemented by manual searching of published bibliographies and abstracts from conferences. Results Early experience with IT ziconotide in clinical trials combined with improved understanding of drug pharmacokinetics in the cerebrospinal fluid have led to a reappraisal of approaches to trialing and initiation of continuous‐infusion therapy in an effort to improve tolerability. The traditional paradigm of trialing by inpatient continuous infusion may be shifting toward outpatient trialing by IT bolus, although definitions of success and specific protocols remain to be agreed upon. Expert consensus on IT continuous infusion with ziconotide suggests a starting dose of 0.5 to 1.2 mcg/day followed by dose titration of ≤0.5 mcg/day on a no more than weekly basis, according to individual patients’ pain reductions and regimen tolerability. Discussion Newer modalities that include patient‐controlled analgesia and nocturnal flex dosing have been shown to hold promise of further improvements in ziconotide efficacy and tolerability. Conclusions Clinical trials and experience confirm the feasibility and usefulness of IT ziconotide in the management of refractory chronic pain. Emerging evidence suggests that additional IT delivery options may further expand the usefulness and benefits of ziconotide.
Collapse
|
31
|
Chuquilin M, Alghalith Y, Fernandez KH. Neurocutaneous disease. J Am Acad Dermatol 2016; 74:197-212. [DOI: 10.1016/j.jaad.2015.04.060] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 04/06/2015] [Accepted: 04/22/2015] [Indexed: 12/14/2022]
|
32
|
Harnessing the knowledge of animal toxins to generate drugs. Pharmacol Res 2016; 112:30-36. [PMID: 26826284 DOI: 10.1016/j.phrs.2016.01.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/07/2016] [Accepted: 01/12/2016] [Indexed: 11/20/2022]
Abstract
Animal toxins present high selectivity and specificity for their molecular targets, and have long been considered as prototypes for developing novel drugs, with some successful cases. In this regard, the variety of molecules found in animal venoms, which can be capable of affecting vital physiological systems, have providing the development of studies focusing on turning those molecules (toxins) into therapeutics to treat several diseases, such as chronic pain, hypertension, thrombosis, cancer, and so on. However, some important issues have been responsible for disrupting the toxin-based drug discovery projects. In this review, we have briefly highlighted the development of drugs based on animal toxins, discussing some successful cases as well as the main causes of failure, pointing out the recent strategies applied to overcome the difficulties related to the translational process in this kind of development scenario.
Collapse
|
33
|
Zamponi GW, Han C, Waxman SG. Voltage-Gated Ion Channels as Molecular Targets for Pain. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
34
|
Zamponi GW. Targeting voltage-gated calcium channels in neurological and psychiatric diseases. Nat Rev Drug Discov 2015; 15:19-34. [DOI: 10.1038/nrd.2015.5] [Citation(s) in RCA: 254] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
35
|
Pessoa BL, Escudeiro G, Nascimento OJM. Emerging Treatments for Neuropathic Pain. Curr Pain Headache Rep 2015; 19:56. [DOI: 10.1007/s11916-015-0530-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
36
|
Navratilova E, Atcherley CW, Porreca F. Brain Circuits Encoding Reward from Pain Relief. Trends Neurosci 2015; 38:741-750. [PMID: 26603560 PMCID: PMC4752429 DOI: 10.1016/j.tins.2015.09.003] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 09/23/2015] [Accepted: 09/24/2015] [Indexed: 01/09/2023]
Abstract
Relief from pain in humans is rewarding and pleasurable. Primary rewards, or reward-predictive cues, are encoded in brain reward/motivational circuits. While considerable advances have been made in our understanding of reward circuits underlying positive reinforcement, less is known about the circuits underlying the hedonic and reinforcing actions of pain relief. We review findings from electrophysiological, neuroimaging, and behavioral studies supporting the concept that the rewarding effect of pain relief requires opioid signaling in the anterior cingulate cortex (ACC), activation of midbrain dopamine neurons, and the release of dopamine in the nucleus accumbens (NAc). Understanding of circuits that govern the reward of pain relief may allow the discovery of more effective and satisfying therapies for patients with acute or chronic pain.
Collapse
Affiliation(s)
- Edita Navratilova
- Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ 85724, USA.
| | | | - Frank Porreca
- Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ 85724, USA; Deparment of Research, Mayo Clinic, Scottsdale, AZ 85453, USA.
| |
Collapse
|
37
|
Patel VB, Wasserman R, Imani F. Interventional Therapies for Chronic Low Back Pain: A Focused Review (Efficacy and Outcomes). Anesth Pain Med 2015; 5:e29716. [PMID: 26484298 PMCID: PMC4604560 DOI: 10.5812/aapm.29716] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 05/17/2015] [Indexed: 01/26/2023] Open
Abstract
CONTEXT Lower back pain is considered to be one of the most common complaints that brings a patient to a pain specialist. Several modalities in interventional pain management are known to be helpful to a patient with chronic low back pain. Proper diagnosis is required for appropriate intervention to provide optimal benefits. From simple trigger point injections for muscular pain to a highly complex intervention such as a spinal cord stimulator are very effective if chosen properly. The aim of this article is to provide the reader with a comprehensive reading for treatment of lower back pain using interventional modalities. EVIDENCE ACQUISITION Extensive search for published literature was carried out online using PubMed, Cochrane database and Embase for the material used in this manuscript. This article describes the most common modalities available to an interventional pain physician along with the most relevant current and past references for the treatment of lower back pain. All the graphics and images were prepared by and belong to the author. RESULTS This review article describes the most common modalities available to an interventional pain physician along with the most relevant current and past references for the treatment of lower back pain. All the graphics and images belong to the author. Although it is beyond the scope of this review article to include a very detailed description of each procedure along with complete references, a sincere attempt has been made to comprehensively cover this very complex and perplexing topic. CONCLUSION Lower back pain is a major healthcare issue and this review article will help educate the pain practitioners about the current evidence based treatment options.
Collapse
Affiliation(s)
- Vikram B. Patel
- Phoenix Interventional Center for Advanced Learning, Algonquin, Illinois, USA
| | - Ronald Wasserman
- Back and Pain Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Farnad Imani
- Pain Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Hayek SM, Hanes MC, Wang C, Veizi IE. Ziconotide Combination Intrathecal Therapy for Noncancer Pain Is Limited Secondary to Delayed Adverse Effects: A Case Series With a 24-Month Follow-Up. Neuromodulation 2015; 18:397-403. [PMID: 25655991 DOI: 10.1111/ner.12270] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/23/2014] [Accepted: 12/04/2014] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The efficacy and safety of ziconotide as a single agent has been evaluated in few short-term clinical trials and open-label studies. Ziconotide use is challenging given its adverse effect (AE) profile. The objective of this study is to describe the long-term efficacy and AEs of ziconotide used as an adjunct to other intrathecal (IT) agents in chronic noncancer pain patients. MATERIALS AND METHODS A case series of chronic noncancer pain patients who had suboptimal pain control from IT therapy. Ziconotide was introduced in the IT infusion mixture after a successful ziconotide trial. Pain scores, IT doses, as well as AEs were recorded and analyzed from trial to initial ziconotide infusion and up to 24 months. RESULTS Fifteen patients underwent ziconotide trials. Four subjects failed the trial, and 11 proceeded to continuous ziconotide treatment. Seven out of 11 patients experienced AEs resulting in ziconotide discontinuation. Two of the seven subjects who required discontinuation of ziconotide had improved pain. Four subjects were able to continue IT ziconotide through 24 months. CONCLUSIONS A high incidence of AEs limits the usefulness of IT ziconotide as adjunct therapy. Our results are limited by the size of our patient population; however, they represent a long follow-up period, which is limited in most current publications on this IT peptide. While ziconotide is a needed IT agent, more studies are necessary to better understand the factors that would improve the treatment to trial ratio as well as the long-term efficacy of IT ziconotide treatment.
Collapse
Affiliation(s)
- Salim M Hayek
- Department of Anesthesiology, Division of Pain Medicine, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, OH, USA
| | - Michael C Hanes
- Department of Anesthesiology, Division of Pain Medicine, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, OH, USA
| | - Connie Wang
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - I Elias Veizi
- Department of Anesthesiology, Division of Pain Medicine, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, OH, USA.,Pain Medicine & Spine Care, Louis Stokes Veterans Administration Medical Center, Cleveland, OH, USA
| |
Collapse
|
39
|
Chen L, Mao J. Update on neuropathic pain treatment: ion channel blockers and gabapentinoids. Curr Pain Headache Rep 2014; 17:359. [PMID: 23888370 DOI: 10.1007/s11916-013-0359-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neuropathic pain is a debilitating chronic pain condition, which remains difficult to treat. The current mainstays of treatment include physical therapy, interventional procedures and medications. Among medications, ion channel blockers and gabapentinoids are the 2 classes of drugs commonly used to treat neuropathic pain. It has been suggested that these medications may be useful to treat a variety of neuropathic pain conditions. This article provides several updates on the utility of both ion channel blockers and gabapentinoids for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Lucy Chen
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
40
|
Bourinet E, Altier C, Hildebrand ME, Trang T, Salter MW, Zamponi GW. Calcium-permeable ion channels in pain signaling. Physiol Rev 2014; 94:81-140. [PMID: 24382884 DOI: 10.1152/physrev.00023.2013] [Citation(s) in RCA: 240] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The detection and processing of painful stimuli in afferent sensory neurons is critically dependent on a wide range of different types of voltage- and ligand-gated ion channels, including sodium, calcium, and TRP channels, to name a few. The functions of these channels include the detection of mechanical and chemical insults, the generation of action potentials and regulation of neuronal firing patterns, the initiation of neurotransmitter release at dorsal horn synapses, and the ensuing activation of spinal cord neurons that project to pain centers in the brain. Long-term changes in ion channel expression and function are thought to contribute to chronic pain states. Many of the channels involved in the afferent pain pathway are permeable to calcium ions, suggesting a role in cell signaling beyond the mere generation of electrical activity. In this article, we provide a broad overview of different calcium-permeable ion channels in the afferent pain pathway and their role in pain pathophysiology.
Collapse
|
41
|
Feldman P, Khanna R. Challenging the catechism of therapeutics for chronic neuropathic pain: Targeting CaV2.2 interactions with CRMP2 peptides. Neurosci Lett 2013; 557 Pt A:27-36. [PMID: 23831344 PMCID: PMC3849117 DOI: 10.1016/j.neulet.2013.06.057] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 06/24/2013] [Accepted: 06/24/2013] [Indexed: 11/25/2022]
Abstract
Chronic neuropathic pain management is a worldwide concern. Pharmaceutical companies globally have historically targeted ion channels as the therapeutic catechism with many blockbuster successes. Remarkably, no new pain therapeutic has been approved by European or American regulatory agencies over the last decade. This article will provide an overview of an alternative approach to ion channel drug discovery: targeting regulators of ion channels, specifically focusing on voltage-gated calcium channels. We will highlight the discovery of an anti-nociceptive peptide derived from a novel calcium channel interacting partner - the collapsin response mediator protein 2 (CRMP2). In vivo administration of this peptide reduces pain behavior in a number of models of neuropathic pain without affecting sympathetic-associated cardiovascular activity, memory retrieval, sensorimotor function, or depression. A CRMP2-derived peptide analgesic, with restricted access to the CNS, represents a completely novel approach to the treatment of severe pain with an improved safety profile. As peptides now represent one of the fastest growing classes of new drugs, it is expected that peptide targeting of protein interactions within the calcium channel complex may be a paradigm shift in ion channel drug discovery.
Collapse
Affiliation(s)
- Polina Feldman
- Sophia Therapeutics LLC, 351 West 10th Street, Indianapolis, IN 46202, USA
| | - Rajesh Khanna
- Sophia Therapeutics LLC, 351 West 10th Street, Indianapolis, IN 46202, USA
- Department of Pharmacology and Toxicology, 635 Barnhill Drive, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, 635 Barnhill Drive, Indianapolis, IN 46202, USA
- Program in Medical Neurosciences, Paul and Carole Stark Neurosciences Research Institute, 950 West Walnut Street, Indianapolis, IN 46202, USA
| |
Collapse
|
42
|
Heifets BD, Smith SM, Leong MS. Acute Cardiovascular Toxicity of Low-Dose Intrathecal Ziconotide. PAIN MEDICINE 2013; 14:1807-9. [DOI: 10.1111/pme.12196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
43
|
Effect of ω-conotoxin MVIIA and Phα1β on paclitaxel-induced acute and chronic pain. Pharmacol Biochem Behav 2013; 114-115:16-22. [PMID: 24148893 DOI: 10.1016/j.pbb.2013.10.014] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 08/31/2013] [Accepted: 10/11/2013] [Indexed: 11/21/2022]
Abstract
The treatment with the chemotherapeutic agent paclitaxel produces a painful peripheral neuropathy, and is associated with an acute pain syndrome in a clinically significant number of patients. However, no standard therapy has been established to manage the acute pain or the chronic neuropathic pain related to paclitaxel. In the present study, we evaluated the analgesic potential of two N-type voltage-gated calcium channel (VGCC) blockers, ω-conotoxin MVIIA and Phα1β, on acute and chronic pain induced by paclitaxel. Adult male rats were treated with four intraperitoneal injections of paclitaxel (1+1+1+1mg/kg, in alternate days) and the development of mechanical hyperalgesia was evaluated 24h (acute painful stage) or 15days (chronic painful stage) after the first paclitaxel injection. Not all animals showed mechanical hyperalgesia 24h after the first paclitaxel injection, but those that showed developed a more intense mechanical hyperalgesia at the chronic painful stage. Intrathecal administration (i.t.) of ω-conotoxin MVIIA (3-300pmol/site) or Phα1β (10-300pmol/site) reduced the mechanical hyperalgesia either at the acute or at the chronic painful stage induced by paclitaxel. When administered at the acute painful stage, ω-conotoxin MVIIA (300pmol/site, i.t.) and Phα1β (300pmol/site, i.t.) prevented the worsening of chronic mechanical hyperalgesia. Furthermore, Phα1β (30-300pmol/site, i.t.) elicited less adverse effects than ω-conotoxin MVIIA (10-300 pmol/site, i.t.). Taken together, our data evidence the involvement of N-type VGCC in pain sensitization induced by paclitaxel and point out the potential of Phα1β as a safer alternative than ω-conotoxin MVIIA to treat the pain related to paclitaxel.
Collapse
|
44
|
O'Connell NE, Wand BM, McAuley J, Marston L, Moseley GL. Interventions for treating pain and disability in adults with complex regional pain syndrome. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2013. [PMID: 23633371 DOI: 10.1002/14651858.cd009416] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND There is currently no strong consensus regarding the optimal management of complex regional pain syndrome although a multitude of interventions have been described and are commonly used. OBJECTIVES To summarise the evidence from Cochrane and non-Cochrane systematic reviews of the effectiveness of any therapeutic intervention used to reduce pain, disability or both in adults with complex regional pain syndrome (CRPS). METHODS We identified Cochrane reviews and non-Cochrane reviews through a systematic search of the following databases: Cochrane Database of Systematic Reviews, Database of Abstracts of Reviews of Effects (DARE), Ovid MEDLINE, Ovid EMBASE, CINAHL, LILACS and PEDro. We included non-Cochrane systematic reviews where they contained evidence not covered by identified Cochrane reviews. The methodological quality of reviews was assessed using the AMSTAR tool.We extracted data for the primary outcomes pain, disability and adverse events, and the secondary outcomes of quality of life, emotional well being and participants' ratings of satisfaction or improvement. Only evidence arising from randomised controlled trials was considered. We used the GRADE system to assess the quality of evidence. MAIN RESULTS We included six Cochrane reviews and 13 non-Cochrane systematic reviews. Cochrane reviews demonstrated better methodological quality than non-Cochrane reviews. Trials were typically small and the quality variable.There is moderate quality evidence that intravenous regional blockade with guanethidine is not effective in CRPS and that the procedure appears to be associated with the risk of significant adverse events.There is low quality evidence that bisphosphonates, calcitonin or a daily course of intravenous ketamine may be effective for pain when compared with placebo; graded motor imagery may be effective for pain and function when compared with usual care; and that mirror therapy may be effective for pain in post-stroke CRPS compared with a 'covered mirror' control. This evidence should be interpreted with caution. There is low quality evidence that local anaesthetic sympathetic blockade is not effective. Low quality evidence suggests that physiotherapy or occupational therapy are associated with small positive effects that are unlikely to be clinically important at one year follow up when compared with a social work passive attention control.For a wide range of other interventions, there is either no evidence or very low quality evidence available from which no conclusions should be drawn. AUTHORS' CONCLUSIONS There is a critical lack of high quality evidence for the effectiveness of most therapies for CRPS. Until further larger trials are undertaken, formulating an evidence-based approach to managing CRPS will remain difficult.
Collapse
Affiliation(s)
- Neil E O'Connell
- Centre for Research in Rehabilitation, School of Health Sciences and Social Care, Brunel University, Uxbridge, UK.
| | | | | | | | | |
Collapse
|
45
|
O'Connell NE, Wand BM, McAuley J, Marston L, Moseley GL. Interventions for treating pain and disability in adults with complex regional pain syndrome. Cochrane Database Syst Rev 2013; 2013:CD009416. [PMID: 23633371 PMCID: PMC6469537 DOI: 10.1002/14651858.cd009416.pub2] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND There is currently no strong consensus regarding the optimal management of complex regional pain syndrome although a multitude of interventions have been described and are commonly used. OBJECTIVES To summarise the evidence from Cochrane and non-Cochrane systematic reviews of the effectiveness of any therapeutic intervention used to reduce pain, disability or both in adults with complex regional pain syndrome (CRPS). METHODS We identified Cochrane reviews and non-Cochrane reviews through a systematic search of the following databases: Cochrane Database of Systematic Reviews, Database of Abstracts of Reviews of Effects (DARE), Ovid MEDLINE, Ovid EMBASE, CINAHL, LILACS and PEDro. We included non-Cochrane systematic reviews where they contained evidence not covered by identified Cochrane reviews. The methodological quality of reviews was assessed using the AMSTAR tool.We extracted data for the primary outcomes pain, disability and adverse events, and the secondary outcomes of quality of life, emotional well being and participants' ratings of satisfaction or improvement. Only evidence arising from randomised controlled trials was considered. We used the GRADE system to assess the quality of evidence. MAIN RESULTS We included six Cochrane reviews and 13 non-Cochrane systematic reviews. Cochrane reviews demonstrated better methodological quality than non-Cochrane reviews. Trials were typically small and the quality variable.There is moderate quality evidence that intravenous regional blockade with guanethidine is not effective in CRPS and that the procedure appears to be associated with the risk of significant adverse events.There is low quality evidence that bisphosphonates, calcitonin or a daily course of intravenous ketamine may be effective for pain when compared with placebo; graded motor imagery may be effective for pain and function when compared with usual care; and that mirror therapy may be effective for pain in post-stroke CRPS compared with a 'covered mirror' control. This evidence should be interpreted with caution. There is low quality evidence that local anaesthetic sympathetic blockade is not effective. Low quality evidence suggests that physiotherapy or occupational therapy are associated with small positive effects that are unlikely to be clinically important at one year follow up when compared with a social work passive attention control.For a wide range of other interventions, there is either no evidence or very low quality evidence available from which no conclusions should be drawn. AUTHORS' CONCLUSIONS There is a critical lack of high quality evidence for the effectiveness of most therapies for CRPS. Until further larger trials are undertaken, formulating an evidence-based approach to managing CRPS will remain difficult.
Collapse
Affiliation(s)
- Neil E O'Connell
- Centre for Research in Rehabilitation, School of Health Sciences and Social Care, Brunel University, Uxbridge, UK.
| | | | | | | | | |
Collapse
|
46
|
Abstract
INTRODUCTION Ziconotide is an N-type calcium channel antagonist to treat chronic pain that is delivered intrathecally. It is the only intrathecal, FDA-approved, non-opioid analgesic and is recommended as first-line therapy. Despite these advantages, a small therapeutic window limits ziconotide's clinical utility, with adverse event (AE) challenges that include, but are not limited to, dizziness, nausea, and somulence. AREAS COVERED Pharmacokinetics, pharmacodynamics, efficacy, safety, trialing, and chronic infusion after searching EMBASE, PubMed, and Cochrane Database of Systemic Reviews were used to search published literature from 1966 to January 1, 2013 to identify studies related to the intrathecal delivery of ziconotide. EXPERT OPINION Ziconotide is a safe and effective strategy to treat chronic pain, although limitations remain, including a small therapeutic window. Low starting doses and slow incremental increases and long titration intervals may improve tolerability. AEs may be mitigated by also employing combination therapy, although further study is needed. Concomitant use of ziconotide and morphine is an option when considering use of FDA-labeled intrathecal drugs in those resistant to monotherapy.
Collapse
Affiliation(s)
- Jason E Pope
- Center for Pain Relief, Inc., 400 Court St, Suite 100, Charleston, WV 25301, USA.
| | | |
Collapse
|
47
|
Navratilova E, Xie JY, King T, Porreca F. Evaluation of reward from pain relief. Ann N Y Acad Sci 2013; 1282:1-11. [PMID: 23496247 DOI: 10.1111/nyas.12095] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The human experience of pain is multidimensional and comprises sensory, affective, and cognitive dimensions. Preclinical assessment of pain has been largely focused on the sensory features that contribute to nociception. The affective (aversive) qualities of pain are clinically significant but have received relatively less mechanistic investigation in preclinical models. Recently, operant behaviors such as conditioned place preference, avoidance, escape from noxious stimulus, and analgesic drug self-administration have been used in rodents to evaluate affective aspects of pain. An important advance of such operant behaviors is that these approaches may allow the detection and mechanistic investigation of spontaneous neuropathic or ongoing inflammatory/nociceptive (i.e., nonevoked) pain that is otherwise difficult to assess in nonverbal animals. Operant measures may allow the identification of mechanisms that contribute differentially to reflexive hypersensitivity or to pain affect and may inform the decision to progress novel mechanisms to clinical trials for pain therapy. Additionally, operant behaviors may allow investigation of the poorly understood mechanisms and neural circuits underlying motivational aspects of pain and the reward of pain relief.
Collapse
Affiliation(s)
- Edita Navratilova
- Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ 85724, USA
| | | | | | | |
Collapse
|
48
|
Obafemi A, Roth B. Prolonged Delirium with Psychotic Features from Omega Conotoxin Toxicity. PAIN MEDICINE 2013; 14:447-8. [DOI: 10.1111/pme.12005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
49
|
Abstract
Neuropathic pain is a clinical entity that presents unique diagnostic and therapeutic challenges. This chapter addresses the classification, epidemiology, pathophysiology, diagnosis, and treatment of neuropathic pain syndrome. Neuropathic pain can be distinguished from nociceptive pain based on clinical signs and symptoms. Although neuropathic pain presents a significant burden to individuals and society, a more accurate assessment of resource utilization, costs, and impairments associated with neuropathic pain would facilitate appropriate planning of healthcare policies. The underlying pathophysiology of neuropathic pain is not well defined. Several theories regarding the mechanism of neuropathic pain have been proposed, including central and peripheral nervous system sensitization, deafferentation, neurogenic inflammation, and the wind up theory. Neuropathic pain is a clinical diagnosis and requires a systematic approach to assessment, including a detailed history, physical examination, and appropriate diagnostic testing. The mainstay of treatment for neuropathic pain is pharmacological, including the use of antidepressants, antiepileptics, topical anesthetics, and opioids. Nonpharmacological treatments include psychological approaches, physical therapy, interventional therapy, spinal cord stimulation, and surgical procedures. Neuropathic pain is difficult to treat, but a combination of therapies may be more effective than monotherapy. Clinical practice guidelines provide an evidence-based approach to the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Eric Kerstman
- Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | | | | | | | | |
Collapse
|
50
|
Khanna R, Wilson SM, Brittain JM, Weimer J, Sultana R, Butterfield A, Hensley K. Opening Pandora's jar: a primer on the putative roles of CRMP2 in a panoply of neurodegenerative, sensory and motor neuron, and central disorders. FUTURE NEUROLOGY 2012; 7:749-771. [PMID: 23308041 DOI: 10.2217/fnl.12.68] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
CRMP2, also known as DPYSL2/DRP2, Unc-33, Ulip or TUC2, is a cytosolic phosphoprotein that mediates axon/dendrite specification and axonal growth. Mapping the CRMP2 interactome has revealed previously unappreciated functions subserved by this protein. Together with its canonical roles in neurite growth and retraction and kinesin-dependent axonal transport, it is now known that CRMP2 interacts with numerous binding partners to affect microtubule dynamics; protein endocytosis and vesicular cycling, synaptic assembly, calcium channel regulation and neurotransmitter release. CRMP2 signaling is regulated by post-translational modifications, including glycosylation, oxidation, proteolysis and phosphorylation; the latter being a fulcrum of CRMP2 functions. Here, the putative roles of CRMP2 in a panoply of neurodegenerative, sensory and motor neuron, and central disorders are discussed and evidence is presented for therapeutic strategies targeting CRMP2 functions.
Collapse
Affiliation(s)
- Rajesh Khanna
- Program in Medical Neurosciences, Paul & Carole Stark Neurosciences Research Institute Indianapolis, IN 46202, USA ; Departments of Pharmacology & Toxicology, Indianapolis, IN 46202, USA ; Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA ; Sophia Therapeutics LLC, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | |
Collapse
|