1
|
López-Carrillo J, Bernáldez-Sarabia J, Pawar TJ, Jiménez S, Dueñas S, Figueroa-Montiel A, Olivares-Romero JL, Granados-Soto V, Licea-Navarro AF, Caram-Salas NL. Systemic antihyperalgesic effect of a novel conotoxin from Californiconus californicus in an inflammatory pain model. FRONTIERS IN PAIN RESEARCH 2025; 5:1500789. [PMID: 39925365 PMCID: PMC11802583 DOI: 10.3389/fpain.2024.1500789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/18/2024] [Indexed: 02/11/2025] Open
Abstract
Introduction This study explores the analgesic potential of the novel conotoxin O1_cal6.4b, derived from Californiconus californicus, as a candidate for pain management in a model of inflammatory pain. Methods O1_cal6.4b was systemically administered to Wistar rats, and its effects on thermal hyperalgesia and motor coordination were evaluated. Comparative analyses were conducted against O1_cal6.4d, ω-MVIIA, and standard analgesics (morphine, dexamethasone, and diclofenac). Structural differences between O1_cal6.4b and O1_cal6.4d were examined using in silico modeling and molecular dynamics simulations. Results Systemic administration of O1_cal6.4b significantly reduced thermal hyperalgesia in a dose-dependent manner without impairing motor coordination. The analgesic effect of O1_cal6.4b was superior to that of O1_cal6.4d, ω-MVIIA, and standard analgesics. Structural analyses revealed notable differences between O1_cal6.4b and O1_cal6.4d, suggesting unique functional properties. Discussion The findings indicate that O1_cal6.4b exhibits a promising analgesic profile with advantages over traditional opioid-based therapies. These results underscore the molecular diversity of conotoxins and highlight their potential as innovative analgesic treatments. Further research is needed to elucidate the mechanism of action of this novel conotoxin.
Collapse
Affiliation(s)
| | | | - Tushar J. Pawar
- Red de Estudios Moleculares Avanzados, Instituto de Ecología (INECOL), Xalapa, Mexico
| | - Samanta Jiménez
- Departamento de Innovación Biomédica, CICESE, Ensenada, Mexico
| | - Salvador Dueñas
- Departamento de Innovación Biomédica, CICESE, Ensenada, Mexico
| | | | | | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Alexei F. Licea-Navarro
- Departamento de Innovación Biomédica, CICESE, Ensenada, Mexico
- Dirección de Impulso a la Innovación y el Desarrollo (DIID), CICESE, Ensenada, Mexico
| | - Nadia L. Caram-Salas
- Departamento de Innovación Biomédica, CICESE, Ensenada, Mexico
- CONAHCYT. Av. Insurgentes Sur 1582, Col. Crédito Constructor, Deleg Benito Juárez, Mexico City, Mexico
| |
Collapse
|
2
|
Deer TR, Hayek SM, Grider JS, Hagedorn JM, McDowell GC, Kim P, Dupoiron D, Goel V, Duarte R, Pilitsis JG, Leong MS, De Andrés J, Perruchoud C, Sukumaran H, Abd-Elsayed A, Saulino M, Patin D, Poree LR, Strand N, Gritsenko K, Osborn JA, Dones I, Bux A, Shah JM, Lindsey BL, Shaw E, Yaksh TL, Levy RM. The Polyanalgesic Consensus Conference (PACC)®: Intrathecal Drug Delivery Guidance on Safety and Therapy Optimization When Treating Chronic Noncancer Pain. Neuromodulation 2024; 27:1107-1139. [PMID: 38752946 DOI: 10.1016/j.neurom.2024.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 10/07/2024]
Abstract
INTRODUCTION The International Neuromodulation Society convened a multispecialty group of physicians and scientists based on expertise with international representation to establish evidence-based guidance on intrathecal drug delivery in treating chronic pain. This Polyanalgesic Consensus Conference (PACC)® project, created more than two decades ago, intends to provide evidence-based guidance for important safety and efficacy issues surrounding intrathecal drug delivery and its impact on the practice of neuromodulation. MATERIALS AND METHODS Authors were chosen on the basis of their clinical expertise, familiarity with the peer-reviewed literature, research productivity, and contributions to the neuromodulation literature. Section leaders supervised literature searches of MEDLINE, BioMed Central, Current Contents Connect, Embase, International Pharmaceutical Abstracts, Web of Science, Google Scholar, and PubMed from 2017 (when PACC® last published guidelines) to the present. Identified studies were graded using the United States Preventive Services Task Force criteria for evidence and certainty of net benefit. Recommendations are based on the strength of evidence or consensus when evidence is scant. RESULTS The PACC® examined the published literature and established evidence- and consensus-based recommendations to guide best practices. Additional guidance will occur as new evidence is developed in future iterations of this process. CONCLUSIONS The PACC® recommends best practices regarding intrathecal drug delivery to improve safety and efficacy. The evidence- and consensus-based recommendations should be used as a guide to assist decision-making when clinically appropriate.
Collapse
Affiliation(s)
- Timothy R Deer
- The Spine and Nerve Centers of the Virginias, Charleston, WV, USA
| | - Salim M Hayek
- Case Western Reserve University, University Hospitals of Cleveland, Cleveland, OH, USA.
| | - Jay S Grider
- UKHealthCare Pain Services, Department of Anesthesiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Jonathan M Hagedorn
- Department of Anesthesiology and Perioperative Medicine, Division of Pain Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Philip Kim
- Christiana Hospital, Newark, DE, USA; Bryn Mawr Hospital, Bryn Mawr, PA, USA
| | - Denis Dupoiron
- Department of Anesthesiology and Pain Medicine, Institut de Cancerologie de L'Ouest, Angers, France
| | - Vasudha Goel
- Department of Anesthesiology, University of Minnesota, Minneapolis, MN, USA
| | - Rui Duarte
- Institute of Population Health, University of Liverpool, Liverpool, UK
| | - Julie G Pilitsis
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | | | - Jose De Andrés
- Anesthesia, Critical Care, and Multidisciplinary Pain Management Department, General University Hospital, València, Spain; Anesthesia Unit, Surgical Specialties Department, Valencia University Medical School, València, Spain
| | | | - Harry Sukumaran
- Department of Anesthesiology, Detroit Medical Center/Wayne State University, Detroit, MI, USA
| | - Alaa Abd-Elsayed
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael Saulino
- Department of Physical Medicine and Rehabilitation, Cooper University Health Care, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Dennis Patin
- University of Miami Health System, Miami, FL, USA
| | - Lawrence R Poree
- Department of Anesthesia and Perioperative Care, University of California at San Francisco, San Francisco, CA, USA
| | - Natalie Strand
- Department of Anesthesiology, Division of Pain Medicine, Mayo Clinic, Phoenix, AZ, USA
| | - Karina Gritsenko
- Department of Anesthesiology, Montefiore Medical Center, Bronx, NY, USA
| | - Jill A Osborn
- St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Ivano Dones
- Department of Neurosurgery, Istituto Nazionale Neurologico "C Besta" of Milan, Milan, Italy
| | - Anjum Bux
- Anesthesia and Chronic Pain Management, Ephraim McDowell Regional Medical Center, Danville, KY, USA
| | - Jay M Shah
- SamWell Institute for Pain Management, Colonia, NJ, USA
| | - Brad L Lindsey
- The Spine and Nerve Centers of the Virginias, Charleston, WV, USA
| | - Erik Shaw
- Shepherd Pain and Spine Institute, Atlanta, GA, USA
| | - Tony L Yaksh
- Anesthesiology and Pharmacology, University of California, San Diego, CA, USA
| | - Robert M Levy
- Neurosurgical Services, Anesthesia Pain Care Consultants, Tamarac, FL, USA
| |
Collapse
|
3
|
Brinzeu A, Berthiller J, Perreton N, Subtil F, Gervaise C, Luaute J, Mertens P. SPIDOL study protocol for the assessment of intrathecal ziconotide antalgic efficacy for severe refractory neuropathic pain due to spinal cord lesions. Trials 2024; 25:595. [PMID: 39244617 PMCID: PMC11380424 DOI: 10.1186/s13063-024-08387-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/06/2024] [Indexed: 09/09/2024] Open
Abstract
RATIONALE Central neuropathic pain resulting from spinal cord injury is notoriously debilitating and difficult to treat with few currently available treatments. A novel molecule with intrathecal administration: Ziconotide has been approved for treatment of refractory neuropathic pain in general. It acts as a presynaptic calcium channel blocker. A pilot study has shown its potential in SCI neuropathic pain patients. OBJECTIVE The aim of this study is to determine the long-term (6 months) efficacy of chronic intrathecal ziconotide for the treatment of neuropathic SCI pain. STUDY DESIGN Multicenter, Randomized, Comparative, Placebo controlled, Double blind clinical trial, with a crossover of random alternated periods of 6 months (placebo or ITZ) for a total of 15 months including a total of 44 patients. STUDY POPULATION • Patients with SCI of various etiologies exhibiting neuropathic pain refractory to non-invasive treatments. • > 18 years. INTERVENTION Intrathecal administration of ziconotide via an implanted pump. STUDY OUTCOMES Primary study outcome Difference in pain intensity for all patients between effective treatment and placebo periods. Secondary study outcomes 1. Continuous evaluation of pain intensity. 2. Percentage of patients with at least 30% of pain reduction. 3. Satisfaction level of the patient pain relief. 4. Declarations of serious adverse events. 5. Duration and intensity of spontaneous and provoked pain. 6. Quality of life. 7. Patient global impression of change. 8. Quantification of daily dosages of analgesic drug intake. 9. Long term memory and neurocognitive effects. 10. Assessment of the patient's physical and emotional distress. NATURE AND EXTENT OF THE BURDEN AND RISKS ASSOCIATED WITH PARTICIPATION, BENEFIT, AND GROUP RELATEDNESS: Participation in this study is in accordance with current treatment protocols for SCI neuropathic pain in France therefore it proposes a treatment that would currently be considered regular practice even though no RCT evidence is yet available. The study gives patients the advantage of directly testing versus placebo a treatment that otherwise entails significant constraints. A Data Safety Monitoring board (DSMB) will be created for continuous safety analysis. Furthermore, patients will be followed in specialized pain centers offering the possibility of continuing their treatment after the study period.
Collapse
Affiliation(s)
- Andrei Brinzeu
- Hospices Civils de Lyon, Lyon, France.
- Department of Neurosurgery, Hopital Neurologique et Neurochirurgical "Pierre Wertheimer" de Lyon, 59 Boulevard Pinel, Lyon, 69003, France.
- Unite de Recherche « Neuropain », CNRS, Universite de Lyon, Lyon, France.
- Neuroscience Research Center, University of Medicine and Pharmacy "Victor Babes" Timisoara, Timisoara, Romania.
| | | | | | | | | | - Jacques Luaute
- Hospices Civils de Lyon, Lyon, France
- Hopital de Reeducation et Readaptation Fonctionelle "Henri Gabrielle", Saint-Genis-Laval, France
| | - Patrick Mertens
- Hospices Civils de Lyon, Lyon, France
- Department of Neurosurgery, Hopital Neurologique et Neurochirurgical "Pierre Wertheimer" de Lyon, 59 Boulevard Pinel, Lyon, 69003, France
- Unite de Recherche « Neuropain », CNRS, Universite de Lyon, Lyon, France
| |
Collapse
|
4
|
Lin J, Chen S, Butt UD, Yan M, Wu B. A comprehensive review on ziconotide. Heliyon 2024; 10:e31105. [PMID: 38779019 PMCID: PMC11110537 DOI: 10.1016/j.heliyon.2024.e31105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Managing severe chronic pain is a challenging task, given the limited effectiveness of available pharmacological and non-pharmacological treatments. This issue continues to be a significant public health concern, requiring a substantial therapeutic response. Ziconotide, a synthetic peptide initially isolated from Conus magus in 1982 and approved by the US Food and Drug Administration and the European Medicines Agency in 2004, is the first-line intrathecal method for individuals experiencing severe chronic pain refractory to other therapeutic measures. Ziconotide produces powerful analgesia by blocking N-type calcium channels in the spinal cord, which inhibits the release of pain-relevant neurotransmitters from the central terminals of primary afferent neurons. However, despite possessing many favorable qualities, including the absence of tolerance development, respiratory depression, and withdrawal symptoms (largely due to the absence of a G-protein mediation mechanism), ziconotide's application is limited due to factors such as intrathecal administration and a narrow therapeutic window resulting from significant dose-related undesired effects of the central nervous system. This review aims to provide a comprehensive and clinically relevant summary of the literatures concerning the pharmacokinetics and metabolism of intrathecal ziconotide. It will also describe strategies intended to enhance clinical efficacy while reducing the incidence of side effects. Additionally, the review will explore the current efforts to refine the structure of ziconotide for better clinical outcomes. Lastly, it will prospect potential developments in the new class of selective N-type voltage-sensitive calcium-channel blockers.
Collapse
Affiliation(s)
- Jinping Lin
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Shuwei Chen
- Fuyang People's Hospital, Hangzhou 311400, China
| | | | - Min Yan
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Bin Wu
- Ocean College, Zhejiang University, Zhoushan 321000, China
| |
Collapse
|
5
|
Ayansiji AO, Gehrke DS, Baralle B, Nozain A, Singh MR, Linninger AA. Determination of spinal tracer dispersion after intrathecal injection in a deformable CNS model. Front Physiol 2023; 14:1244016. [PMID: 37817986 PMCID: PMC10561273 DOI: 10.3389/fphys.2023.1244016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/30/2023] [Indexed: 10/12/2023] Open
Abstract
Background: Traditionally, there is a widely held belief that drug dispersion after intrathecal (IT) delivery is confined locally near the injection site. We posit that high-volume infusions can overcome this perceived limitation of IT administration. Methods: To test our hypothesis, subject-specific deformable phantom models of the human central nervous system were manufactured so that tracer infusion could be realistically replicated in vitro over the entire physiological range of pulsating cerebrospinal fluid (CSF) amplitudes and frequencies. The distribution of IT injected tracers was studied systematically with high-speed optical methods to determine its dependence on injection parameters (infusion volume, flow rate, and catheter configurations) and natural CSF oscillations in a deformable model of the central nervous system (CNS). Results: Optical imaging analysis of high-volume infusion experiments showed that tracers spread quickly throughout the spinal subarachnoid space, reaching the cervical region in less than 10 min. The experimentally observed biodispersion is much slower than suggested by the Taylor-Aris dispersion theory. Our experiments indicate that micro-mixing patterns induced by oscillatory CSF flow around microanatomical features such as nerve roots significantly accelerate solute transport. Strong micro-mixing effects due to anatomical features in the spinal subarachnoid space were found to be active in intrathecal drug administration but were not considered in prior dispersion theories. Their omission explains why prior models developed in the engineering community are poor predictors for IT delivery. Conclusion: Our experiments support the feasibility of targeting large sections of the neuroaxis or brain utilizing high-volume IT injection protocols. The experimental tracer dispersion profiles acquired with an anatomically accurate, deformable, and closed in vitro human CNS analog informed a new predictive model of tracer dispersion as a function of physiological CSF pulsations and adjustable infusion parameters. The ability to predict spatiotemporal dispersion patterns is an essential prerequisite for exploring new indications of IT drug delivery that targets specific regions in the CNS or the brain.
Collapse
Affiliation(s)
- Ayankola O. Ayansiji
- Department of Bioengineering, University of Illinois Chicago, Chicago, IL, United States
- Department of Chemical Engineering, University of Illinois Chicago, Chicago, IL, United States
| | - Daniel S. Gehrke
- Department of Bioengineering, University of Illinois Chicago, Chicago, IL, United States
| | - Bastien Baralle
- UIC Student Intern From EPF, Ecole D’Ingénieur, Paris, France
| | - Ariel Nozain
- UIC Student Intern From EPF, Ecole D’Ingénieur, Paris, France
| | - Meenesh R. Singh
- Department of Chemical Engineering, University of Illinois Chicago, Chicago, IL, United States
| | - Andreas A. Linninger
- Department of Bioengineering, University of Illinois Chicago, Chicago, IL, United States
- Department of Neurosurgery, University of Illinois Chicago, Chicago, IL, United States
| |
Collapse
|
6
|
Yaksh TL, Santos GGD, Borges Paes Lemes J, Malange K. Neuraxial drug delivery in pain management: An overview of past, present, and future. Best Pract Res Clin Anaesthesiol 2023; 37:243-265. [PMID: 37321769 DOI: 10.1016/j.bpa.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 06/17/2023]
Abstract
Activation of neuraxial nociceptive linkages leads to a high level of encoding of the message that is transmitted to the brain and that can initiate a pain state with its attendant emotive covariates. As we review here, the encoding of this message is subject to a profound regulation by pharmacological targeting of dorsal root ganglion and dorsal horn systems. Though first shown with the robust and selective modulation by spinal opiates, subsequent work has revealed the pharmacological and biological complexity of these neuraxial systems and points to several regulatory targets. Novel therapeutic delivery platforms, such as viral transfection, antisense and targeted neurotoxins, point to disease-modifying approaches that can selectively address the acute and chronic pain phenotype. Further developments are called for in delivery devices to enhance local distribution and to minimize concentration gradients, as frequently occurs with the poorly mixed intrathecal space. The field has advanced remarkably since the mid-1970s, but these advances must always address the issues of safety and tolerability of neuraxial therapy.
Collapse
Affiliation(s)
- Tony L Yaksh
- Department of Anesthesiology University of California, San Diego, San Diego CA, 92103, USA.
| | | | | | - Kaue Malange
- Department of Anesthesiology University of California, San Diego, San Diego CA, 92103, USA
| |
Collapse
|
7
|
De Andres J, Hayek S, Perruchoud C, Lawrence MM, Reina MA, De Andres-Serrano C, Rubio-Haro R, Hunt M, Yaksh TL. Intrathecal Drug Delivery: Advances and Applications in the Management of Chronic Pain Patient. FRONTIERS IN PAIN RESEARCH 2022; 3:900566. [PMID: 35782225 PMCID: PMC9246706 DOI: 10.3389/fpain.2022.900566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/02/2022] [Indexed: 11/19/2022] Open
Abstract
Advances in our understanding of the biology of spinal systems in organizing and defining the content of exteroceptive information upon which higher centers define the state of the organism and its role in the regulation of somatic and automatic output, defining the motor response of the organism, along with the unique biology and spatial organization of this space, have resulted in an increased focus on therapeutics targeted at this extracranial neuraxial space. Intrathecal (IT) drug delivery systems (IDDS) are well-established as an effective therapeutic approach to patients with chronic non-malignant or malignant pain and as a tool for management of patients with severe spasticity and to deliver therapeutics that address a myriad of spinal pathologies. The risk to benefit ratio of IDD makes it a useful interventional approach. While not without risks, this approach has a significant therapeutic safety margin when employed using drugs with a validated safety profile and by skilled practioners. The present review addresses current advances in our understanding of the biology and dynamics of the intrathecal space, therapeutic platforms, novel therapeutics, delivery technology, issues of safety and rational implementation of its therapy, with a particular emphasis upon the management of pain.
Collapse
Affiliation(s)
- Jose De Andres
- Surgical Specialties Department, Valencia University Medical School, Valencia, Spain
- Anesthesia Critical Care and Pain Management Department, Valencia, Spain
- *Correspondence: Jose De Andres
| | - Salim Hayek
- Department of Anesthesiology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Christophe Perruchoud
- Pain Center and Department of Anesthesia, La Tour Hospital, Geneva, Switzerland
- Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Melinda M. Lawrence
- Department of Anesthesiology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Miguel Angel Reina
- Department of Anesthesiology, Montepríncipe University Hospital, Madrid, Spain
- CEU-San-Pablo University School of Medicine, Madrid, Spain
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, United States
- Facultad de Ciencias de la Salud Universidad Francisco de Vitoria, Madrid, Spain
| | | | - Ruben Rubio-Haro
- Anesthesia and Pain Management Department, Provincial Hospital, Castellon, Spain
- Multidisciplinary Pain Clinic, Vithas Virgen del Consuelo Hospital, Valencia, Spain
| | - Mathew Hunt
- Department of Physiology, Karolinska Institute, Stockholm, Sweden
| | - Tony L. Yaksh
- Departments of Anesthesiology and Pharmacology, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
8
|
Leino T, Viitamaa T, Salonen JS, Pesonen U, Haapalinna A. Effects of fadolmidine, an α 2 -adrenoceptor agonist, as an adjuvant to spinal bupivacaine on antinociception and motor function in rats and dogs. Pharmacol Res Perspect 2021; 9:e00830. [PMID: 34302721 PMCID: PMC8308519 DOI: 10.1002/prp2.830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 11/06/2022] Open
Abstract
α2 -Adrenoceptor agonists such as clonidine and dexmedetomidine are used as adjuvants to local anesthetics in regional anesthesia. Fadolmidine is an α2 -adrenoceptor agonist developed especially as a spinal analgesic. The current studies investigate the effects of intrathecally administered fadolmidine with a local anesthetic, bupivacaine, on antinociception and motor block in conscious rats and dogs. The antinociceptive effects of intrathecal fadolmidine and bupivacaine alone or in combination were tested in the rat tail-flick and the dog's skin twitch models. The durations of motor block in rats and in dogs were also assessed. In addition, the effects on sedation, mean arterial blood pressure, heart rate, respiratory rate and body temperature were evaluated in telemetrized dogs. Concentrations of fadolmidine in plasma and spinal cord were determined after intrathecal and intravenous administration in rats. Co-administration of intrathecal fadolmidine with bupivacaine increased the magnitude and duration of the antinociceptive effects and prolonged motor block without hypotension. The interaction of the antinociceptive effect was synergistic in its nature in rats. Concentration of fadolmidine in plasma was very low after intrathecal dosing. Taken together, these studies show that fadolmidine as an adjuvant to intrathecal bupivacaine provides enhanced sensory-motor block and enables a reduction of the doses of both drugs. The results indicate that co-administration of fadolmidine with intrathecal bupivacaine was able to achieve an enhanced antinociceptive effect without hypotension and could thus represent a suitable combination for spinal anesthesia.
Collapse
Affiliation(s)
- Tiina Leino
- Orion Corporation Orion PharmaR&DTurkuFinland
| | | | | | - Ullamari Pesonen
- Integrative Physiology and Pharmacology Research UnitInstitute of BiomedicineFaculty of MedicineUniversity of TurkuTurkuFinland
| | | |
Collapse
|
9
|
Bienfait F, Jubier-Hamon S, Seegers V, Pluchon YM, Lebrec N, Jaoul V, Boré F, Delorme T, Robert J, Bellanger M, Sorrieul J, Dupoiron D. First Evaluation Switching From Ropivacaine to Highly Concentrated Bupivacaine in Intrathecal Mixtures for Cancer Pain. Neuromodulation 2021; 24:1215-1222. [PMID: 34181790 DOI: 10.1111/ner.13469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/23/2021] [Accepted: 05/17/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Intrathecal drug delivery is widely used for intractable cancer pain treatment. A combination of drugs with morphine and bupivacaine is recommended in first line therapy. In France, we use ropivacaine 10 mg/mL instead of bupivacaine 5 mg/mL, the only concentration available. Bupivacaine 40 mg/mL has been available in France only since July 2020 under temporary authorization of use. OBJECTIVES The main objective of the study was to evaluate the safety, efficacy by pain assessment, to analyze drug dosage changes, to report adverse events (AEs) and conversion ratios switching from ropivacaine to bupivacaine. Secondary objective was to evaluate costs differences. MATERIALS AND METHODS We conducted this retrospective follow-up monocentric study within the Institut de Cancérologie de l'Ouest (ICO) Pain Department in Angers, France. We included 14 patients aged 18 years and above, implanted with an Intrathecal Drug Delivery Systems (IDDS) for cancer pain treatment and followed up at ICO from July 2020 to February 2021 after switching from ropivacaine to bupivacaine. We used a continuous infusion mode and Bolus could be added through Personal Therapy Manager (PTM). RESULTS The median conversion ratio between ropivacaine and bupivacaine was 0.68 (0.65; 0.69) and resulted in no significant change in numeric rating scale evaluation (p = 0.10). We observed moderate and rapidly reversible AEs such as clinical hypotension (29%) and motor block after bolus (21%). The estimated median hospital cost per day was significantly lower (p = 0.05) for the bupivacaine refills than for the last ropivacaine pump refill, decreasing from US$ 61.7 (49.6; 70.5) to US$ 50.4 (45.9; 60.4). The median reimbursement per day from the National Health Insurance (NHI) was three times lower for bupivacaine pump refill when compared to the last ropivacaine pump refill (p < 0.01), decreasing from US$ 179.10 (156.79; 182.91) to US$ 64.59 (59.85; 71.89). CONCLUSION Switching from ropivacaine to bupivacaine in IDDS appears more efficacious while remaining just as secure, and at lower cost.
Collapse
Affiliation(s)
- Florent Bienfait
- Department of Anesthesiology and Pain Medicine, Institut de Cancerologie de l'Ouest, Angers, France
| | - Sabrina Jubier-Hamon
- Department of Anesthesiology and Pain Medicine, Institut de Cancerologie de l'Ouest, Angers, France
| | - Valérie Seegers
- Department of Epidemiology and Biostatistics, Institut de Cancerologie De l'Ouest, Angers, France
| | | | - Nathalie Lebrec
- Department of Anesthesiology and Pain Medicine, Institut de Cancerologie de l'Ouest, Angers, France
| | - Virginie Jaoul
- Department of Anesthesiology and Pain Medicine, Institut de Cancerologie de l'Ouest, Angers, France
| | - François Boré
- Department of Anesthesiology and Pain Medicine, Institut de Cancerologie de l'Ouest, Angers, France
| | - Thierry Delorme
- Department of Anesthesiology and Pain Medicine, Institut de Cancerologie de l'Ouest, Angers, France
| | - Julien Robert
- Pharmacy, Institut de Cancerologie de l'Ouest, Angers, France
| | - Martine Bellanger
- Department of Human and Social Sciences, Institut de Cancerologie de l'Ouest, Nantes, France.,EHESP School of Public Health, Paris, France
| | - Jérémy Sorrieul
- Pharmacy, Institut de Cancerologie de l'Ouest, Angers, France
| | - Denis Dupoiron
- Department of Anesthesiology and Pain Medicine, Institut de Cancerologie de l'Ouest, Angers, France
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
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
|
13
|
Schmidt-Rondon E, Wang Z, Malkmus SA, Di Nardo A, Hildebrand K, Page L, Yaksh TL. Effects of opioid and nonopioid analgesics on canine wheal formation and cultured human mast cell degranulation. Toxicol Appl Pharmacol 2018; 338:54-64. [PMID: 29111148 PMCID: PMC9841896 DOI: 10.1016/j.taap.2017.10.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/10/2017] [Accepted: 10/20/2017] [Indexed: 02/06/2023]
Abstract
Mast cell (MC) degranulation has been implicated in the side effect profile of a variety of clinically useful agents. Thus, after intrathecal delivery, formation of space-occupying, meningeally-derived masses may be related to local MC degranulation. We systematically characterized degranulating effects of opioid and nonopioid analgesics on cutaneous flares in the dog and in primary human MC (hMC) cultures. METHODS Dogs were anesthetized with IV propofol and received intradermal (ID) injections (50μL). Flare diameters were measured at 30min. Drugs showing flare responses were tested after intramuscular (IM) cromolyn (10mg/kg), a MC stabilizer. Human primary MCs (human cord blood CD34+/CD45+ cells) were employed and β-hexosaminidase in cell-free supernatants were measured to assess degranulation. RESULTS A significant skin flare for several classes of agents was observed including opioids, ziconotide, ketamine, ST-91, neostigmine, adenosine, bupivacaine, lidocaine, MK-801 and 48/80. Tizanidine, fentanyl, alfentanil, gabapentin and baclofen produced no flare. Flare produced by all ID agents, except adenosine, bupivacaine and lidocaine, was reduced by cromolyn. Naloxone had no effect upon opiate or 48/80 evoked flares. In hMC studies, 48/80 resulted in a concentration-dependent release of β-hexosaminidase. The rank order of drug-induced hMC β-hexosaminidase release was similar to that for flares. CONCLUSIONS A variety of therapeutically useful drugs degranulate MCs. This action may account for side effects such as the intrathecal granuloma resulting from spinally-delivered opioids. This degranulating effect may be useful in predicting potential intrathecal toxicity in the development of novel agents.
Collapse
Affiliation(s)
- Eric Schmidt-Rondon
- Department of Anesthesiology, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States
| | - Zhenping Wang
- Department of Dermatology, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States
| | - Shelle A. Malkmus
- Department of Anesthesiology, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States
| | - Anna Di Nardo
- Department of Dermatology, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States
| | - Keith Hildebrand
- Medtronic, Inc., Neuromodulation, 7000 Central Avenue NE, RCE470, Minneapolis, MN 55432, United States
| | - Linda Page
- Medtronic, Inc., Neuromodulation, 7000 Central Avenue NE, RCE470, Minneapolis, MN 55432, United States
| | - Tony L. Yaksh
- Department of Anesthesiology, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States,Corresponding author at: Department of Anesthesiology 0818, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093-0818, United States, (T.L. Yaksh)
| |
Collapse
|
14
|
Pope JE, Deer TR, Amirdelfan K, McRoberts WP, Azeem N. The Pharmacology of Spinal Opioids and Ziconotide for the Treatment of Non-Cancer Pain. Curr Neuropharmacol 2017; 15:206-216. [PMID: 26861471 PMCID: PMC5412702 DOI: 10.2174/1570159x14666160210142339] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 04/01/2015] [Accepted: 02/05/2016] [Indexed: 12/29/2022] Open
Abstract
Background Intrathecal drug delivery has undergone a revitalization following a better understanding of this delivery route and its pharmacokinetics. Driven by patient safety and outcomes, clinicians are motivated to rethink the traditional spinal infusion pump patient selection criteria and indications. We review the current understanding of the pharmacology of commonly employed intrathecal agents and the clinical relevance. Methods Search strategies for data acquisition included Medline database, PubMed, Google scholar, along with international and national professional meeting content, with key words including pharmacology of opioids, intrathecal therapy, ziconotide, pharmacokinetics, and intrathecal drug delivery. The search results were limited to the English language. Results Over 300 papers were identified. The literature was condensed and digested to evaluate the most commonly used medications in practice, sto serve as a foundation for review. We review on-label medications: ziconotide and morphine, and off label medications including fentanyl, sufentail, and hydromorphine. Conclusion Intrathecal therapy has level-one evidence for use for malignant pain and nonmalignant pain, with continued cost savings and improved safety. To most effectively serve our patients, a clear appreciation for the pharmacology of these commonly employed medication is paramount..
Collapse
Affiliation(s)
- J E Pope
- Center for Pain Relief, Charleston, WV, USA. United States
| | | | | | | | | |
Collapse
|
15
|
Deer TR, Pope JE, Hayek SM, Bux A, Buchser E, Eldabe S, De Andrés JA, Erdek M, Patin D, Grider JS, Doleys DM, Jacobs MS, Yaksh TL, Poree L, Wallace MS, Prager J, Rauck R, DeLeon O, Diwan S, Falowski SM, Gazelka HM, Kim P, Leong M, Levy RM, McDowell II G, McRoberts P, Naidu R, Narouze S, Perruchoud C, Rosen SM, Rosenberg WS, Saulino M, Staats P, Stearns LJ, Willis D, Krames E, Huntoon M, Mekhail N. The Polyanalgesic Consensus Conference (PACC): Recommendations on Intrathecal Drug Infusion Systems Best Practices and Guidelines. Neuromodulation 2017; 20:96-132. [DOI: 10.1111/ner.12538] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 12/13/2022]
Affiliation(s)
| | | | | | - Anjum Bux
- Anesthesia and Chronic Pain Management; Ephraim McDowell Regional Medical Center; Danville KY USA
| | - Eric Buchser
- Anaesthesia and Pain Management Department; EHC Hosptial, Morges, and CHUV University Hospital; Lausanne Switzerland
| | - Sam Eldabe
- The James Cook University Hospital; Middlesbrough UK
| | - Jose A. De Andrés
- Valencia School of Medicine; Hospital General Universitario; Valencia Spain
| | - Michael Erdek
- Anesthesiology and Critical Care Medicine; Johns Hopkins University School of Medicine; Baltimore MD USA
| | | | - Jay S. Grider
- University of Kentucky College of Medicine, UK HealthCare Pain Services; Lexington KY USA
| | | | | | - Tony L. Yaksh
- Anesthesiology and Pharmacology; University of California; San Diego CA USA
| | - Lawrence Poree
- Pain Clinic of Monterey Bay, University of California at San Francisco; San Francisco CA USA
| | | | - Joshua Prager
- Center for the Rehabilitation Pain Syndromes (CRPS) at UCLA Medical Plaza; Los Angeles CA USA
| | - Richard Rauck
- Carolina Pain Institute, Wake Forest Baptist Health; Winston-Salem NC USA
| | - Oscar DeLeon
- Roswell Park Cancer Institute, SUNY; Buffalo NY USA
| | - Sudhir Diwan
- Manhattan Spine and Pain Medicine; Lenox Hill Hospital; New York NY USA
| | | | | | - Philip Kim
- Bryn Mawr Hospital; Bryn Mawr PA, USA
- Christiana Hospital; Newark DE USA
| | | | | | | | | | - Ramana Naidu
- San Francisco Medical Center, University of California; San Francisco CA USA
| | - Samir Narouze
- Summa Western Reserve Hospital; Cuyahoga Falls OH USA
| | | | | | | | | | - Peter Staats
- Premier Pain Management Centers; Shrewsbury NJ, USA
- Johns Hopkins University; Baltimore MD USA
| | | | | | - Elliot Krames
- Pacific Pain Treatment Center (ret.); San Francisco CA USA
| | - Marc Huntoon
- Vanderbilt University Medical Center; Nashville TN USA
| | | |
Collapse
|
16
|
Deer TR, Hayek SM, Pope JE, Lamer TJ, Hamza M, Grider JS, Rosen SM, Narouze S, Perruchoud C, Thomson S, Russo M, Grigsby E, Doleys DM, Jacobs MS, Saulino M, Christo P, Kim P, Huntoon EM, Krames E, Mekhail N. The Polyanalgesic Consensus Conference (PACC): Recommendations for Trialing of Intrathecal Drug Delivery Infusion Therapy. Neuromodulation 2017; 20:133-154. [PMID: 28042906 DOI: 10.1111/ner.12543] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 06/20/2016] [Accepted: 07/06/2016] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Intrathecal (IT) drug infusion is an appropriate and necessary tool in the algorithm to treat refractory cancer and noncancer pain. The decision-making steps/methodology for selecting appropriate patients for implanted targeted drug delivery systems is controversial and complicated. Therefore, a consensus on best practices for determining appropriate use of IT drug infusion may involve testing/trialing this therapy before implantation. METHODS This current Polyanalgesic Consensus Conference (PACC) update was designed to address the deficiencies and emerging innovations since the previous PACC convened in 2012. A literature search identified publications available since the previous PACC publications in 2014, and relevant sources were contributed by the PACC members. After reviewing the literature, the panel determined the evidence levels and degrees of recommendations. The developed consensus was ranked as strong (>80%), moderate (50-79%), or weak (<49%). RESULTS The trialing for IT drug delivery systems (IDDS) remains an area of continued controversy. The PACC recommendations for trialing are presented in 34 consensus points and cover trialing for morphine, ziconotide, and medication admixtures; starting doses and titration practices; measurements of success; trial settings and monitoring; management of systemic opioids during trialing; and the role of psychological evaluation. Finally, the PACC describes clinical scenarios in which IT trialing is required or not required. CONCLUSION The PACC provides consensus guidance on best practices of trialing for IDDS implants. In addition, the PACC recommends that no trial may be required in certain patient populations.
Collapse
Affiliation(s)
| | - Salim M Hayek
- Hospitals Case Medical Center, Case Western Reserve University, Cleveland, OH, USA
| | | | | | - Maged Hamza
- Virginia Commonwealth University Spine Center, Richmond, VA, USA
| | - Jay S Grider
- UK HealthCare Pain Services, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Steven M Rosen
- Delaware Valley Pain & Spine Institute, Chalfront, PA, USA
| | | | | | - Simon Thomson
- Basildon and Thurrock University Hospitals FHT, Essex, UK
| | - Marc Russo
- Hunter Pain Clinic, Newcastle, NSW, Australia
| | | | | | | | | | | | - Philip Kim
- Bryn Mawr Hospital, Bryn Mawr, PA, USA.,Christiana Hospital, Newark, DE, USA
| | | | - Elliot Krames
- Pacific Pain Treatment Center (ret.), San Francisco, CA, USA
| | - Nagy Mekhail
- Cleveland Clinic, Evidence-Based Pain Management Research, Cleveland, OH, USA
| |
Collapse
|
17
|
Ziconotide intrathecal delivery as treatment for secondary therapeutic failure of motor cortex stimulation after 6 years. Neurochirurgie 2016; 62:284-288. [PMID: 27771111 DOI: 10.1016/j.neuchi.2016.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 03/21/2016] [Accepted: 06/12/2016] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Motor cortex stimulation is a well-known treatment modality for refractory neuropathic pain. Nevertheless, some cases of therapeutic failure have been described but alternative therapies for these cases are rarely reported. CASE REPORT The patient presented with neuropathic pain in his right arm due to a cervical syrinx which was surgically treated by a shunt in 2003 with no clinical improvement. As alternative therapy, after an evaluation by repetitive magnetic transcranial stimulation with significant benefit, motor cortex stimulation was successfully implanted in 2004. In 2010, a similar pain occurred in the same territory. Local mean pain visual analogical scale (VAS) increased to 82/100. A newer generation stimulation device was then implanted and, within a period of 8months, different stimulation parameter settings were tested, without any pain relief. An intrathecal drug delivery pump was then implanted in 2011, and the upper extremity catheter was located at the cervicothoracic junction. There was no postoperative complication. A bitherapy was initiated at a daily dosage of 0.2mg morphine and 1.3μg ziconotide, not modified since August 2013. At 43months follow-up, mean VAS was 21/100 with improvement of daily life and spare-time activities, anxiety and depression, quality of life (as measured by the SF-36 survey and EQ5D-3L questionnaire). DISCUSSION Refractory neuropathic pain treated by motor cortex stimulation may be considered in palliative situations, and secondary therapeutic failure offers only a few perspectives. Intrathecal ziconotide, indicated as a first-line drug in non-cancer pain, could be proposed in such cases. CONCLUSION Intrathecal drug delivery including ziconotide in refractory neuropathic pain represents a reasonable option with a good clinical tolerance.
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Targeted intrathecal drug delivery systems (IDDS) are an option in algorithms for the treatment of patients with moderate-to-severe chronic refractory pain. This article is intended to review the literature regarding IDDS published over the last year, with special attention to the Polyanalgesic Consensus Conference 2012. RECENT FINDINGS The recommendations made by the Polyanalgesic Consensus Conference 2012 are reviewed. Separate considerations of intrathecal drug therapy for neuropathic and nociceptive pain syndromes and the new concept of 'microdosing' are discussed in this article. SUMMARY This review includes the recommendations for the use of IDDS, trialing, and recent reports of complications (especially, the occurrence of granulomas). In addition, the latest documents on cerebrospinal fluid and potential lines of future development are discussed.
Collapse
|
19
|
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
|
20
|
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
|
21
|
Pope JE, Deer TR. Intrathecal drug delivery for pain: a clinical guide and future directions. Pain Manag 2016; 5:175-83. [PMID: 25971641 DOI: 10.2217/pmt.15.12] [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] [Indexed: 11/21/2022] Open
Abstract
Intrathecal infusion of medications allows for a direct delivery of agents to the receptors in which they act. This method of treatment is indicated in severe chronic pain of cancer or noncancer origin. In recent years, the use of these devices has increased and the general pain physician, referring doctor and concerned parties such as insurers, needs a better understanding of safety and efficacy. This article is an overview of this therapeutic option and also provides an update on new innovations and forward thinking approaches at improving patient selection and appropriateness of use.
Collapse
Affiliation(s)
- Jason E Pope
- Summit Pain Alliance, 392 Tesconi Court, Santa Rosa, CA 95401 USA
| | | |
Collapse
|
22
|
Pope JE, Deer TR, McRoberts WP. Intrathecal Therapy: The Burden of Being Positioned as a Salvage Therapy. PAIN MEDICINE 2015; 16:2036-8. [DOI: 10.1111/pme.12782] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 02/08/2015] [Accepted: 03/29/2015] [Indexed: 02/06/2023]
|
23
|
Bäckryd E, Sörensen J, Gerdle B. Ziconotide Trialing by Intrathecal Bolus Injections: An Open-Label Non-Randomized Clinical Trial in Postoperative/Posttraumatic Neuropathic Pain Patients Refractory to Conventional Treatment. Neuromodulation 2015; 18:404-13. [PMID: 25879804 DOI: 10.1111/ner.12293] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 02/19/2015] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The aim of this open-label, non-randomized, clinical trial was to evaluate the feasibility of trialing ziconotide by intrathecal bolus injections. MATERIAL AND METHODS Twenty-three patients, who had peripheral neuropathic pain refractory to pharmacological treatment and were under consideration for Spinal Cord Stimulation, received up to three ziconotide bolus injections according to a comprehensive algorithm. After a first injection of 2.5 μg, the patients progressed in the algorithm depending on the presence or absence of pain reduction and significant adverse events. A patient was considered a "responder" if experiencing pain reduction and no significant adverse event on two consecutive occasions at the same dosage. RESULTS We found a low proportion of responders (13%). However 30% of patients experienced ≥30% pain reduction on a least one injection, yielding a number needed to treat of ∼3 for clinically significant pain relief. Pain intensity changed significantly over time (0-6 h) (p = 0.047) after a mean ziconotide dose of 2.75 μg. Adverse events were as expected, and no serious adverse event occurred. We did not find any statistical association between response to Spinal Cord Stimulation and response to ziconotide. CONCLUSIONS Ziconotide bolus injection trialing seems feasible, but the proportion of responders in the present study was low. Adverse events were as expected, and no serious adverse event occurred. The predictive power of ziconotide bolus trialing remains unclear, and the pharmacological profile of ziconotide (slow tissue penetration due to high hydrophilicity) calls the rationale for bolus trialing into question.
Collapse
Affiliation(s)
- Emmanuel Bäckryd
- Rehabilitation Medicine, Department of Medicine and Health Sciences, Linköping University, Linköping, SE 581 85, Sweden.,Pain and Rehabilitation Centre, UHL, County Council of Östergötland, Linköping, SE 581 85, Sweden
| | - Jan Sörensen
- Rehabilitation Medicine, Department of Medicine and Health Sciences, Linköping University, Linköping, SE 581 85, Sweden.,Pain and Rehabilitation Centre, UHL, County Council of Östergötland, Linköping, SE 581 85, Sweden
| | - Björn Gerdle
- Rehabilitation Medicine, Department of Medicine and Health Sciences, Linköping University, Linköping, SE 581 85, Sweden.,Pain and Rehabilitation Centre, UHL, County Council of Östergötland, Linköping, SE 581 85, Sweden
| |
Collapse
|
24
|
Ren Z, Wang L, Qin M, You Y, Pan W, Zhou L, Sun D, Xu A. Pharmacological characterization of conotoxin lt14a as a potent non-addictive analgesic. Toxicon 2015; 96:57-67. [PMID: 25617597 DOI: 10.1016/j.toxicon.2015.01.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 01/19/2015] [Accepted: 01/21/2015] [Indexed: 01/18/2023]
Abstract
Conotoxin lt14a is a small peptide consisting of 13 amino acids. It was originally identified from the cDNA of Conus litteratus in the South China Sea. Previous reports showed lt14a exhibited antinociceptive activity using a hot plate-induced pain mouse model and acted as an antagonist of neuronal nicotinic acetylcholine receptors. We confirmed that conotoxin lt14a administration resulted in antinociception activity using a mouse inflammatory pain model and a rat model of mechanically-induced pain. The mRNA expression of c-fos and NOS in the spinal cord of rats was suppressed by lt14a. Labeling of lt14a with an Alexa Fluor 488 ester showed that lt14a was bound to the surface of PC12 cells and that this binding was inhibited by pre-application of the nicotinic acetylcholine receptor (nAChR) antagonist tubocurarine chloride (TUB) and the nAChR blocker hexamethonium bromide (HB). These data confirm previous reports that showed lt14a binds to the surface of PC12 cells via nAChRs with patch clamp whole-cell recordings. Additional results showed that lt14a suppressed extracellular signal-regulated kinase (ERK1/2) phosphorylation in PC12 cells activated by Ach. Our results showed that lt14a did not induce drug dependence but rather suppressed morphine withdrawal symptoms. Our work suggests that lt14a is a novel antinociceptive agent that targets the nAChR receptor without inducing drug dependence.
Collapse
Affiliation(s)
- Zhenghua Ren
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, National Engineering Research Center of South China Sea Marine Biotechnology, Department of Biochemistry, College of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China
| | - Lei Wang
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, National Engineering Research Center of South China Sea Marine Biotechnology, Department of Biochemistry, College of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China.
| | - Mengying Qin
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, National Engineering Research Center of South China Sea Marine Biotechnology, Department of Biochemistry, College of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China
| | - Yuwen You
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, National Engineering Research Center of South China Sea Marine Biotechnology, Department of Biochemistry, College of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China
| | - Wuguang Pan
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, National Engineering Research Center of South China Sea Marine Biotechnology, Department of Biochemistry, College of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China
| | - Liang Zhou
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, National Engineering Research Center of South China Sea Marine Biotechnology, Department of Biochemistry, College of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China
| | - Dandan Sun
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, National Engineering Research Center of South China Sea Marine Biotechnology, Department of Biochemistry, College of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China
| | - Anlong Xu
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, National Engineering Research Center of South China Sea Marine Biotechnology, Department of Biochemistry, College of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China; Beijing University of Chinese Medicine, 11 Bei San Huan Dong Road, Beijing, 100029, People's Republic of China.
| |
Collapse
|
25
|
Pope JE, Deer TR. Intrathecal Pharmacology Update: Novel Dosing Strategy for Intrathecal Monotherapy Ziconotide on Efficacy and Sustainability. Neuromodulation 2015; 18:414-20. [PMID: 25708382 DOI: 10.1111/ner.12274] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 12/29/2014] [Accepted: 12/30/2014] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Intrathecal drug delivery is a well-defined strategy to treat malignant and nonmalignant pain. Ziconotide is a well-studied intrathecal medicine option that has many attractive qualities, as it is non-granulomagenic, overdose or underdose is not associated with cardiopulmonary compromise or death, and is a non-opoid analgesic. However, it has had slow adoption into pain care algorithms because it has been historically plagued with the connotation of having a narrow therapeutic window and a low sustainability rate. We introduce a novel dosing strategy to improve patient outcomes and sustainability. METHODS Patients were identified as being an intrathecal candidate and trialed with ziconotide based on the current standard of care. Patient demographics, diagnosis, previous treatment failures, and pre-implant visual analog scale (VAS) scores were recorded. Once the trial was deemed successful, based on the dual bolusing strategy, the patient underwent device implantation. Consecutive patients were prospectively followed. Ziconotide was then initiated with a flex dosing strategy, weighted during nocturnal dosing. Outcome endpoints included: reduction in VAS, side effects, durability of therapy, and systemic opioid use prior to implant and at last visit were noted (calculated to daily morphine equivalents). Primary endpoint was tolerability of ziconotide at three months following new dosing strategy. No industry support or funding was obtained for this project. RESULTS All enrolled patients met the endpoint of the study of tolerability of ziconotide at three months. Numbers declined to 75% of patients at four months, and 70% of patients at six months. The discontinuing side-effects were most commonly urinary retention and visual hallucinations. There were no serious adverse events and no unresolved complications reported. Numerical rating scale (NRS) decreased on average from 9.06 to 1.8. Opioid reduction in morphine equivalents averaged 91.5% DISCUSSION The efficacy and tolerability of monotherapy ziconotide may be improved by using a weighted bolus flex dosing strategy as compared with slow continuous infusions. CONCLUSION We present a novel strategy to deliver ziconotide using a unique continuous infusion flex dosing strategy. Further randomized, prospective, higher-powered studies are needed to critically evaluate the conclusions suggested by this limited prospective case series.
Collapse
Affiliation(s)
- Jason E Pope
- Center for Pain Relief, Teays Valley, WV, USA
- Center for Pain Relief, Charleston, WV, USA
| | | |
Collapse
|
26
|
Berecki G, Daly NL, Huang YH, Vink S, Craik DJ, Alewood PF, Adams DJ. Effects of arginine 10 to lysine substitution on ω-conotoxin CVIE and CVIF block of Cav2.2 channels. Br J Pharmacol 2015; 171:3313-27. [PMID: 24628243 DOI: 10.1111/bph.12686] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 02/28/2014] [Accepted: 03/05/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE ω-Conotoxins CVIE and CVIF (CVIE&F) selectively inhibit Cav2.2 channels and are lead molecules in the development of novel analgesics. At physiological membrane potentials, CVIE&F block of Cav2.2 channels is weakly reversible. To improve reversibility, we designed and synthesized arginine CVIE&F analogues in which arginine was substituted for lysine at position 10 ([R10K]CVIE&F), and investigated their serum stability and pharmacological actions on voltage-gated calcium channels (VGCCs). EXPERIMENTAL APPROACH Changes in peptide structure due to R10K substitution were assessed by NMR. Peptide stability in human serum was analysed by reversed-phase HPLC and MS over a 24 h period. Two-electrode voltage-clamp and whole-cell patch clamp techniques were used to study [R10K]CVIE&F effects on VGCC currents in Xenopus oocytes and rat dorsal root ganglion neurons respectively. KEY RESULTS R10K substitution did not change the conserved ω-conotoxin backbone conformations of CVIE&F nor the ω-conotoxin selectivity for recombinant or native Cav2.2 channels, although the inhibitory potency of [R10K]CVIF was better than that of CVIF. At -80 mV, the R10K chemical modification significantly affected ω-conotoxin-channel interaction, resulting in faster onset kinetics than those of CVIE&F. Heterologous and native Cav2.2 channels recovered better from [R10K]CVIE&F block than CVIE&F. In human serum, the ω-conotoxin half-lives were 6-10 h. CVIE&F and [R10K]CVIE&F were more stable than CVID. CONCLUSIONS AND IMPLICATIONS R10K substitution in CVIE&F significantly alters the kinetics of ω-conotoxin action and improves reversibility without diminishing conotoxin potency and specificity for the Cav2.2 channel and without diminishing the serum stability. These results may help generate ω-conotoxins with optimized kinetic profiles for target binding.
Collapse
Affiliation(s)
- G Berecki
- Health Innovations Research Institute, RMIT University, Melbourne, Vic, Australia
| | | | | | | | | | | | | |
Collapse
|
27
|
Horazeck C, Huh AS, Huh BK. Acute Rhabdomyolysis in a Patient with Long-Term Exposure to Intrathecal Ziconotide: A Case Report. Pain Pract 2015; 15:E34-9. [DOI: 10.1111/papr.12273] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 11/02/2014] [Indexed: 01/04/2023]
Affiliation(s)
- Christian Horazeck
- Department of Anesthesiology; Duke University Medical Center; Durham North Carolina U.S.A
| | | | - Billy K. Huh
- Department of Pain Medicine; The University of Texas MD Anderson Cancer Center; Houston Texas U.S.A
| |
Collapse
|
28
|
High cerebrospinal fluid levels of interleukin-10 attained by AAV in dogs. Gene Ther 2014; 22:202-8. [PMID: 25354684 DOI: 10.1038/gt.2014.96] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 07/23/2014] [Accepted: 08/22/2014] [Indexed: 12/15/2022]
Abstract
Intrathecal (IT) gene transfer using adeno-associated virus (AAV) may be clinically promising as a treatment for chronic pain if it can produce sufficiently high levels of a transgene product in the cerebrospinal fluid (CSF). Although this strategy was developed in rodents, no studies investigating CSF levels of an analgesic or antiallodynic protein delivered by IT AAV have been performed in large animals. Interleukin-10 (IL-10) is an antiallodynic cytokine for which target therapeutic levels have been established in rats. The present study tested IT AAV8 encoding either human IL-10 (hIL-10) or enhanced green fluorescent protein (EGFP) in a dog model of IT drug delivery. AAV8/hIL-10 at a dose of 3.5 × 10(12) genome copies induced high hIL-10 levels in the CSF, exceeding the target concentration previously found to be antiallodynic in rodents by >1000-fold. AAV8/EGFP targeted the primary sensory and motor neurons and the meninges. hIL-10, a xenogeneic protein in dogs, induced anti-hIL-10 antibodies detectable in the CSF and serum of dogs. The high hIL-10 levels demonstrate the efficacy of AAV for delivery of secreted transgenes into the IT space of large animals, suggesting a strong case for further development toward clinical testing.
Collapse
|
29
|
De Andres J, Asensio-Samper JM, Fabregat-Cid G. Intrathecal delivery of analgesics. Methods Mol Biol 2014; 1141:249-78. [PMID: 24567144 DOI: 10.1007/978-1-4939-0363-4_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Targeted intrathecal (IT) drug delivery systems (IDDS) are an option in algorithms for the treatment of patients with moderate to severe chronic refractory pain when more conservative options fail. This therapy is well established and supported by several publications. It has shown efficacy and is an important tool for the treatment of spasticity, and both cancer and nonmalignant pain. Recent technological advances, new therapeutic applications, reported complications, and the costs as well as maintenance required for this therapy require the need to stay up-to-date about new recommendations that may improve outcomes. This chapter reviews all technological issues regarding IDDS implantation with follow-up, and pharmacological recommendations published during recent years that provide evidence-based decision making process in the management of chronic pain and spasticity in patients.
Collapse
Affiliation(s)
- Jose De Andres
- Multidisciplinary Pain Management Department, Valencia University General Hospital, Valencia, Spain
| | | | | |
Collapse
|
30
|
Wiese AJ, Rathbun M, Butt MT, Malkmus SA, Richter PJ, Osborn KG, Xu Q, Veesart SL, Steinauer JJ, Higgins D, Lappi DA, Russell B, Yaksh TL. Intrathecal substance P-saporin in the dog: distribution, safety, and spinal neurokinin-1 receptor ablation. Anesthesiology 2013; 119:1163-77. [PMID: 24051388 DOI: 10.1097/aln.0b013e3182a95164] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Neurokinin-1 receptors (NK1-rs) located on superficial dorsal horn neurons are essential for integration of nociceptive input. Intrathecal injection of substance P-saporin (SP-SAP) leads to local loss of spinal NK1-r (+) neurons suggesting its potential as a therapeutic agent for chronic pain. The authors determined, in a canine model, effects of lumbar intrathecal SP-SAP. METHODS Distribution of SP-SAP and Saporin was determined in plasma, lumbar cerebrospinal fluid, and tissue. Safety of intrathecal SP-SAP was determined in four groups (six dogs each) administered 0 (0.9% saline), 1.5, 15, or 150 µg SP-SAP through lumbar intrathecal catheters. Behavioral, physiologic, and biochemical variables were assessed. Spinal tissues were collected at 7 and approximately 90 days, or earlier if significant morbidity developed, and analyzed for NK1-r (+) neuron loss and histopathology. RESULTS SP-SAP and Saporin were detectable in lumbar cerebrospinal fluid for up to 4 and 24 h, respectively. Animals receiving intrathecal saline, 1.5, or 15 µg of SP-SAP showed no persistent neurologic deficits. Three animals receiving 150 µg of SP-SAP developed pelvic limb paraparesis and were euthanized prematurely. Immunohistochemistry and in situ hybridization cell counts confirmed a significant reduction in NK1-r (+) in superficial dorsal horn neurons from lumbar spinal cord after intrathecal administration of 15 and 150 µg of SP-SAP. A significant loss of NK1-r neurons in the lumbar ventral horn occurred only with 150-µg SP-SAP. CONCLUSION Intrathecal 15-µg SP-SAP reduced dorsal, but not ventral, NK1-r (+) neurons at the spinal level of delivery with minimal side effects, whereas 150-µg SP-SAP resulted in motor neuron toxicity.
Collapse
Affiliation(s)
- Ashley J Wiese
- * Research Fellow, † Staff Research Associate, # Postdoctoral Fellow, §§ Professor of Anesthesiology and Pharmacology, Department of Anesthesiology, University of California, San Diego, California. ‡ President, Tox Path Specialists, LLC, Frederick, Maryland. § Director, Campus Veterinary Medicine, ‖ Associate Director, Diagnostic Laboratory, Office of Animal Research, University of California. ** Vice President, †† President/CSO, ‡‡ Research Scientist, Advanced Targeting Systems, San Diego, California
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Role of meningeal mast cells in intrathecal morphine-evoked granuloma formation. Anesthesiology 2013; 118:664-78. [PMID: 23426209 DOI: 10.1097/aln.0b013e31828351aa] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Intrathecal morphine forms granulomas that arise from the adjacent arachnoid membrane. The authors propose that these inflammatory cells exit the meningeal vasculature secondary to meningeal mast cell degranulation. METHODS Three sets of experiments were accomplished in dogs: (1) ex vivo meningeal mast cell degranulation (histamine release was measured ex vivo from canine dura incubated with opiates); (2) in vivo cutaneous mast cell degranulation (flare areas on the dog abdomen were measured after subcutaneous opiates); and (3) in vivo granuloma pharmacology. Dogs with lumbar intrathecal catheters received infusion of intrathecal saline or intrathecal morphine. Intrathecal morphine dogs received (1) no other treatment (control); (2) twice-daily subcutaneous naltrexone; (3) intrathecal co-infusion of cromolyn; or (4) twice-daily subcutaneous cromolyn for the 24- to 28-day study course. RESULTS Morphine but not fentanyl evoked dural histamine release, which was blocked by cromolyn but not naloxone. Wheal/flare was produced by subcutaneous morphine, methadone, hydromorphone, but not fentanyl, and was unaffected by naltrexone but prevented by cromolyn. Granulomas occurred in all dogs receiving intrathecal morphine (15 of 15); subcutaneous naltrexone had no effect on granulomas (six of six) but was reduced by concurrent intrathecal cromolyn (zero of five) or twice-daily subcutaneous cromolyn (one of five). CONCLUSIONS The pharmacology of cutaneous/dural mast cell degranulation and intrathecal granulomas are comparable, not mediated by opioid receptors, and reduced by agents preventing mast cell degranulation. If an agent produces cutaneous mast cell degranulation at concentrations produced by intrathecal delivery, the agent may initiate granulomas.
Collapse
|
32
|
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
|
33
|
Pleticha J, Maus TP, Jeng-Singh C, Marsh MP, Al-Saiegh F, Christner JA, Lee KH, Beutler AS. Pig lumbar spine anatomy and imaging-guided lateral lumbar puncture: a new large animal model for intrathecal drug delivery. J Neurosci Methods 2013; 216:10-5. [PMID: 23518340 DOI: 10.1016/j.jneumeth.2013.03.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 03/08/2013] [Accepted: 03/11/2013] [Indexed: 12/22/2022]
Abstract
Intrathecal (IT) administration is an important route of drug delivery, and its modelling in a large animal species is of critical value. Although domestic swine is the preferred species for preclinical pharmacology, no minimally invasive method has been established to deliver agents into the IT space. While a "blind" lumbar puncture (LP) can sample cerebrospinal fluid (CSF), it is unreliable for drug delivery in pigs. Using computed tomography (CT), we determined the underlying anatomical reasons for this irregularity. The pig spinal cord was visualised terminating at the S2-S3 level. The lumbar region contained only small amounts of CSF found in the lateral recess. Additional anatomical constraints included ossification of the midline ligaments, overlapping lamina with small interlaminar spaces, and a large bulk of epidural adipose tissue. Accommodating the the pig CT anatomy, we developed a lateral LP (LLP) injection technique that employs advanced planning of the needle path and monitoring of the IT injection progress. The key features of the LLP procedure involved choosing a vertebral level without overlapping lamina or spinal ligament ossification, a needle trajectory crossing the midline, and entering the IT space in its lateral recess. Effective IT delivery was validated by the injection of contrast media to obtain a CT myelogram. LLP represents a safe and reliable method to deliver agents to the lumbar pig IT space, which can be implemented in a straightforward way by any laboratory with access to CT equipment. Therefore, LLP is an attractive large animal model for preclinical studies of IT therapies.
Collapse
Affiliation(s)
- Josef Pleticha
- Department of Anesthesiology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Yaksh TL, Steinauer JJ, Veesart SL, Malkmus SA. Alfentanil: correlations between absence of effect upon subcutaneous mast cells and absence of granuloma formation after intrathecal infusion in the dog. Neuromodulation 2012; 16:459-66; discussion 466. [PMID: 23170763 DOI: 10.1111/j.1525-1403.2012.00534.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 09/20/2012] [Accepted: 09/28/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND We hypothesize that intrathecal (IT) granulomas arising from the IT infusion of several opiates may result from the degranulation of meningeal mast cells (MC). Given functional covariance between cutaneous and meningeal MC, we propose that opioids that do not degranulate cutaneous MC will not produce a granuloma. An opioid meeting this criteria is the phenylpiperadine alfentanil HCl. METHODS Three experiments were accomplished in dogs. 1) Cutaneous MC degranulation. Flare areas on the dog abdomen were measured after intradermal alfentanil, morphine, or compound 48-80. 2) Dose ranging of analgesic effects of IT alfentanil infusion. Dogs with lumbar IT catheters received continuous infusion for 24 hours of different concentrations (1-20 mg/mL/d) of alfentanil and analgesic effects were assessed. 3) Granuloma inducing effects. Dogs received IT alfentanil (20 mg/mL/d; N = 5; 22-28 days) or morphine (12 mg/mL/d; N = 3; 22-30 days) and spinal cord harvested for histopathology after 22-30 days of infusion. RESULTS 1) Intradermal morphine (10 mg/mL) and compound 48-80 (1 mg/mL) but not alfentanil at concentrations up to 20 mg/mL produced a cutaneous flare. IT alfentanil infusion produced increases in thermal escape latency at concentrations as low as 2 mg/mL/day. A significant depression of arousal was noted in the dogs receiving 20 mg/mL. Over the 22- to 30-day infusion period, morphine (12 mg/mL/day) resulted in granulomas in all three animals examined whereas IT alfentanil at 20 mg/mL/day failed to initiate a granuloma in any animal. CONCLUSIONS These results support the hypothesis linking MC degranulation and IT granulomas.
Collapse
Affiliation(s)
- Tony L Yaksh
- Anesthesiology Research, Department of Anesthesiology, University of California, San Diego, CA, USA
| | | | | | | |
Collapse
|