Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
BMC Anesthesiol ; 22(1): 401, 2022 12 24.
Article in English | MEDLINE | ID: mdl-36564723

ABSTRACT

BACKGROUND: Alfaxalone is a fast acting intravenous anaesthetic with high therapeutic index. It is an analogue of the naturally-occurring neurosteroid allopregnanolone responsible for maintenance of cognition and neuroprotection by activation of brain pregnane X receptors and consequent increased production of mature brain-derived neurotrophic factor (m-BDNF). Two studies are reported here: an in vitro study investigated whether alfaxalone activates human pregnane X receptors (h-PXR) as effectively as allopregnanolone; and a clinical study that measured postoperative changes in serum m-BDNF and cognition in patients after alfaxalone anaesthesia compared with propofol and sevoflurane. METHODS: In vitro Activation of h-PXR by allopregnanolone and alfaxalone solutions (206 - 50,000 nM) was measured using human embryonic kidney cells expressing h-PXR hybridised and linked to the firefly luciferase gene. Light emission by luciferase stimulated by each ligand binding with h-PXR was measured. Clinical A double blind prospective randomised study of patients undergoing hip arthroplasty anaesthetised with alfaxalone TIVA (n = 8) or propofol TIVA (n = 3) or propofol plus sevoflurane inhalational anaesthesia (n = 4). The doses of anaesthetics were titrated to the same depth of anaesthesia (BIS 40-60). Subjects' cognitive performance was assessed using the Grooved Pegboard Test, Digit Symbol Substitution Test (DSST) and Mini Mental State examination (MMSE) for 7 days postoperatively. Serum m-BDNF concentrations were measured for 7 postoperative days. RESULTS: In vitro Allopregnanolone and alfaxalone both activated h-PXR, alfaxalone being more efficacious than allopregnanolone: 50,000 nM, p = 0.0019; 16,700 nM, p = 0.0472; 5600 nM, p = 0.0031. Clinical Alfaxalone treated subjects scored better than propofol and sevoflurane anaesthetised patients in the cognition tests: (MMSE p = 0.0251; Grooved Pegboard test dominant hand pre v post anaesthesia scores p = 0.8438 for alfaxalone and p = 0.0156 for propofol and propofol/sevoflurane combined). The higher cognition scores were accompanied by higher serum m-BDNF levels in the alfaxalone anaesthetised patients (p < 0.0001). CONCLUSIONS: These results suggest that sedation and anaesthesia induced by the synthetic neuroactive steroid alfaxalone may be accompanied by effects normally caused by physiological actions of allopregnanolone at PXR, namely, increased secretion of m-BDNF and consequent neuroprotection and preservation of cognition. TRIAL REGISTRATION: The clinical trial was registered on 17/01/2018 with the Australian New Zealand Clinical Trials Registry: registration number ACTRN12618000064202 [Universal Trial Number U1111-1198-0412].


Subject(s)
Anesthetics, Inhalation , Propofol , Humans , Propofol/pharmacology , Sevoflurane/pharmacology , Brain-Derived Neurotrophic Factor , Prospective Studies , Pregnanolone/pharmacology , Australia , Anesthetics, Intravenous , Cognition , Anesthesia, Inhalation/methods , Double-Blind Method
2.
Anesth Analg ; 130(3): 704-714, 2020 03.
Article in English | MEDLINE | ID: mdl-31124836

ABSTRACT

BACKGROUND: Previous formulations of alfaxalone have shown it to be a fast-acting intravenous anesthetic with high therapeutic index. Alfaxalone has been reformulated for human use as Phaxan, an aqueous solution of 10 mg/mL of alfaxalone and 13% betadex. This study assessed the pharmacokinetic (PK) and pharmacodynamic (PD) characteristics of alfaxalone given as a bolus intravenous injection of this formulation to human male volunteers. METHODS: A dose of 0.5 mg/kg (0.42-0.55 mg/kg) of alfaxalone [mean (range)] was given by single intravenous bolus injection to 12 healthy subjects. Plasma alfaxalone concentrations and bispectral index (BIS) values were analyzed using an integrated pharmacokinetic-pharmacodynamic (PKPD) model using nonlinear mixed-effects models. Effect (BIS) was described using a sigmoidal fractional maximum effect (EMAX) model. All parameters were scaled using allometry and standardized to a 70-kg person using exponents of 0.75 for clearance parameters (CL, Q2, and Q3), 1.0 for volumes (V1, V2, and V3), and 0.25 for time-related parameters half-time keo (t1/2keo). RESULTS: A 3-compartment model used to fit PK data with an additional compartment, linked by t1/2keo to describe the effect compartment, yielded alfaxalone PK parameter estimates: CL: 1.08 L/min; 0.87-1.34 L/min (median; 95% confidence interval [CI]); central volume of distribution (V1): 0.99 L; 0.53-2.05 L (median; 95% CI); intercompartment CLs (Q2): 0.87 L/min; 0.32-1.71 L/min (median; 95% CI) and Q3: 0.46 L/min; 0.19-1.03 L/min (median; 95% CI); and peripheral volumes of distribution (V2): 6.36 L; 2.79-10.7 L (median; 95% CI) and V3: 19.1 L; 8.61-37.4 L (median; 95% CI). PD interrogation assumed a baseline BIS of 96, with an estimated EMAX: 0.94; 0.71-0.99 (median; 95% CI), a plasma concentration (Cp) for 50% effect (C50): 0.98 mg/L; 0.83-1.09 mg/L (median; 95% CI), and a Hill coefficient (γ): 12.1; 6.7-15 (median; 95% CI). The t1/2keo was 8 minutes; 4.70-12.8 minutes (median; 95% CI). The mean time to a BIS 50 was 0.94 minutes (standard deviation [SD] = 0.2 minutes). CONCLUSIONS: After a single bolus intravenous injection, alfaxalone has a high plasma CL equal to hepatic blood flow as reported for earlier studies of bolus injections of a previous formulation of alfaxalone. The plasma levels associated with BIS values of <60 are comparable to those previously reported in patients anesthetized with alfaxalone. The t1/2keo is relatively high, but the large Hill coefficient contributes to rapid onset and offset of action. This information can inform future studies of this formulation.


Subject(s)
Anesthetics/administration & dosage , Anesthetics/pharmacokinetics , Consciousness/drug effects , Pregnanediones/administration & dosage , Pregnanediones/pharmacokinetics , Adolescent , Adult , Anesthetics/blood , Consciousness Monitors , Drug Compounding , Half-Life , Healthy Volunteers , Humans , Injections, Intravenous , Male , Metabolic Clearance Rate , Models, Biological , New Zealand , Pregnanediones/blood , Young Adult
3.
Anesth Analg ; 121(4): 914-924, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26226029

ABSTRACT

BACKGROUND: Phaxan™ (PHAX, Chemic Labs, Canton, MA) is an aqueous solution of 10 mg/mL alphaxalone and 13% 7-sulfobutylether ß-cyclodextrin (betadex). In preclinical studies, PHAX is a fast onset-offset IV anesthetic like propofol, but causes less cardiovascular depression. This first-in-man study was designed to find the anesthetic dose of PHAX and to compare it with an equivalent dose of propofol for safety, efficacy, and quality of recovery from anesthesia and sedation. METHODS: The study adhered to compliance with Good Clinical Practices regulations (clinical trials registry number, ACTRN12611000343909). This randomized, double-blind study compared PHAX and propofol using a Bayesian algorithm to determine dose equivalence for effects on the bispectral index (BIS). Male volunteers, ASA physical status I, gave written informed consent (n = 12 per group; PHAX or propofol). Parameters assessed for 80 minutes after drug injection (single bolus dose) were pain on injection, involuntary movement, BIS, blood pressure, need for airway support, and, as measures of recovery from sedation, the Richmond Agitation and Sedation Scale and the Digit Symbol Substitution Test. Arterial blood was withdrawn for biochemistry, hematology, and complement levels. RESULTS: No subject complained of pain on injection with PHAX, whereas 8 of the 12 subjects given propofol did. Nine PHAX and 8 propofol subjects reached BIS values of ≤50: median (interquartile range [IQR]) mg/kg dose = 0.5 (0.5-0.6) for PHAX and 2.9 (2.4-3.0) for propofol. The lowest median BIS reached was 27 to 28 for both agents with no significant differences between them for timing of onset and recovery of BIS. The concomitant median changes in systolic and diastolic blood pressures were -11% vs -19% for systolic and -25% vs -37% for diastolic in PHAX- and propofol-treated subjects, respectively. Nine of the 12 propofol-treated subjects and none of 12 PHAX-treated subjects required airway support. For subjects reaching an equivalent BIS of ≤50: a Richmond Agitation and Sedation Scale score of 0 was reached at a median of 5 (IQR, 5-10) and 15 (IQR, 10-20) minutes after PHAX and propofol, respectively; BIS returned to 90 at a mean of 21 (SD, 10.1) and 21 (SD, 9.2) minutes after PHAX and propofol, respectively; and Digit Symbol Substitution Test scores returned to predrug injection values at median of 50 (IQR, 35-72.5) and 42.5 (IQR, 35-76.3) minutes after PHAX and propofol, respectively. There was no increase in C3 and C4 complement fractions after either drug. CONCLUSIONS: PHAX causes fast-onset, short-duration anesthesia with fast cognitive recovery similar to propofol, but with less cardiovascular depression, or airway obstruction and no pain on injection.


Subject(s)
Anesthesia/methods , Anesthetics, Intravenous/administration & dosage , Hypnotics and Sedatives/administration & dosage , Pregnanediones/administration & dosage , Propofol/administration & dosage , Water/administration & dosage , Adolescent , Adult , Anesthetics, Intravenous/adverse effects , Blood Pressure/drug effects , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Double-Blind Method , Humans , Hypnotics and Sedatives/adverse effects , Male , Pregnanediones/adverse effects , Propofol/adverse effects , Water/adverse effects , Young Adult
4.
Pain Med ; 16(9): 1781-93, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25800174

ABSTRACT

BACKGROUND: "Burst ketamine" (BK) is the long-term infusion of subanesthetic ketamine in combination with an opioid. It is used clinically with mixed success to provide long-term pain relief and improve opioid response in patients. BK has not been simulated preclinically, therefore, its effectiveness was investigated in an animal model of neuropathic pain--streptozotocin-induced diabetic neuropathy. METHODS: Diabetic neuropathic rats were randomized to receive a subcutaneous infusion of ketamine 20 mg/kg/day plus morphine 20 mg/kg/day (BK), either drug alone at the same dose, or sham treatment. Drugs were administered continuously over 5 days via osmotic minipump. Antihyperalgesic effects and antinociceptive responsiveness to morphine (0.625-10 mg/kg, i.p.) were assessed at 2, 4, 6, and 12 weeks post-treatment using paw withdrawal latency (PWL) from noxious heat (thermal hyperalgesia) and mechanical touch (tactile allodynia). RESULTS: Antihyperalgesic effects with significant increases in PWL from noxious heat occurred following BK and ketamine-only infusion, persisting 12 and 4 weeks, respectively. Opioid-sparing effects from noxious heat with increased sensitivity to morphine analgesia also occurred for 6 weeks after BK and 2 weeks after ketamine treatment; acute treatment with the maximum nonsedating dose of morphine (5 mg/kg) produced an antinociceptive effect in these two groups, but not in sham-treated rats. In morphine-only infusion rats, hyperalgesia and opioid insensitivity were both increased. CONCLUSIONS: This is the first preclinical study to use a model of neuropathic pain to demonstrate the utility of the BK procedure for delivering a long-lasting reduction in hyperalgesia and improved antinociceptive responsiveness to opioids.


Subject(s)
Analgesics/administration & dosage , Diabetic Neuropathies/drug therapy , Ketamine/administration & dosage , Morphine/administration & dosage , Neuralgia/drug therapy , Animals , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Drug Therapy, Combination , Hyperalgesia/drug therapy , Infusions, Subcutaneous , Male , Neuralgia/etiology , Rats , Rats, Wistar
5.
Anesth Analg ; 120(5): 1025-1031, 2015 May.
Article in English | MEDLINE | ID: mdl-25517195

ABSTRACT

BACKGROUND: Alphaxalone is a neuroactive steroid anesthetic that is poorly water soluble. It was formulated in 1972 as Althesin® using Cremophor® EL, a nonionic surfactant additive. The product was a versatile short-acting IV anesthetic used in clinical practice in many countries from 1972 to 1984. It was withdrawn from clinical practice because of hypersensitivity to Cremophor EL. In the investigations reported here, we compared the properties of 3 anesthetics: a new aqueous solution of alphaxalone dissolved in 7-sulfobutyl-ether-ß-cyclodextrin (SBECD, a water-soluble molecule with a lipophilic cavity that enables drug solubilization in water); a Cremophor EL preparation of alphaxalone; and propofol. METHODS: Two solutions of alphaxalone (10 mg/mL) were prepared: one using 13% w/v solution of SBECD in 0.9% saline (PHAX) and the other a solution of alphaxalone prepared as described in the literature using 20% Cremophor EL (ALTH). A solution of propofol (10 mg/mL; PROP) in 10% v/v soya bean oil emulsion was used as a comparator anesthetic. Jugular IV catheters were implanted in male Wistar rats (180-220 g) under halothane anesthesia. Separate groups of 10 implanted rats each were given IV injections of PHAX, ALTH, or PROP from 1.2 mg/kg to lethal doses. Doses of each drug that caused anesthesia (loss of righting reflex and response to tail pinch) and lethality in 50% of rats were calculated by probit analysis. The drugs were also compared for effects on arterial blood pressure and heart rate. RESULTS: IV PHAX, ALTH, and PROP caused dose-related sedation and anesthesia, with 50% effective dose (ED50) values for loss of righting reflex being 2.8, 3.0, and 4.6 mg/kg, respectively. PROP led to death in 10 of 10 rats at doses >30 mg/kg (50% lethal dose (LD50) = 27.7 mg/kg). A dose of alphaxalone 53 mg/kg as ALTH caused 10 of 10 rats to die (LD50 = 43.6 mg/kg), whereas none died when given the same doses of alphaxalone formulated in SBECD. PHAX caused 20% lethality at the maximal dose tested of 84 mg/kg. PHAX caused less cardiovascular depression than PROP. Control experiments with the 3 drug-free vehicles showed no effects. CONCLUSIONS: Alphaxalone caused fast-onset anesthesia at the same dose for both formulations (PHAX and ALTH). The use of SBECD as a drug-solubilizing excipient did not alter the anesthetic effect of alphaxalone, but it did increase the therapeutic index of alphaxalone in PHAX compared with ALTH. PHAX has a higher safety margin than the propofol lipid formulation and also the alphaxalone formulation in Cremophor EL (ALTH).


Subject(s)
Anesthetics, Intravenous/pharmacology , Excipients/chemistry , Glycerol/analogs & derivatives , Pregnanediones/pharmacology , Propofol/pharmacology , Water/chemistry , beta-Cyclodextrins/chemistry , Anesthetics, Intravenous/chemistry , Anesthetics, Intravenous/toxicity , Animals , Arterial Pressure/drug effects , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Glycerol/chemistry , Heart Rate/drug effects , Lethal Dose 50 , Male , Motor Activity/drug effects , Pain Threshold/drug effects , Pregnanediones/chemistry , Pregnanediones/toxicity , Propofol/chemistry , Propofol/toxicity , Rats, Wistar , Reflex/drug effects , Risk Assessment , Sleep/drug effects , Solubility , Soybean Oil/chemistry , Time Factors
6.
Pain Med ; 13(11): 1444-56, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23078152

ABSTRACT

OBJECTIVE: Current treatments for cancer pain are often inadequate, particularly when metastasis to bone is involved. The addition to the treatment regimen of another drug that has a complementary analgesic effect may increase the overall analgesia without the necessity to increase doses, thus avoiding dose-related side effects. This project investigated the synergistic effect of the addition of the potassium channel (KCNQ2-3) modulator flupirtine to morphine treatment in a rat model of prostate cancer-induced bone pain. DESIGN: Syngeneic prostate cancer cells were injected into the right tibia of male Wistar rats under anesthesia. This led to expanding tumor within the bone in 2 weeks, together with the concurrent development of hyperalgesia to noxious heat. Paw withdrawal thresholds from noxious heat were measured before and after the maximum non-sedating doses of morphine and flupirtine given alone and in combinations. Dose-response curves for morphine (0.13-5.0 mg/kg ip) and flupirtine (1.25-10.0 mg/kg ip) given alone and in fixed-dose combinations were plotted and subjected to an isobolographic analysis. RESULTS: Both morphine (ED50 = 0.74 mg/kg) and flupirtine (ED50 = 3.32 mg/kg) caused dose-related anti-hyperalgesia at doses that did not cause sedation. Isobolographic analysis revealed that there was a synergistic interaction between flupirtine and morphine. Addition of flupirtine to morphine treatment improved morphine anti-hyperalgesia, and resulted in the reversal of cancer-induced heat hyperalgesia. CONCLUSIONS: These results suggest that flupirtine in combination with morphine may be useful clinically to provide better analgesia at lower morphine doses in the management of pain caused by tumors growing in bone.


Subject(s)
Aminopyridines/administration & dosage , Analgesics/administration & dosage , Bone Neoplasms/drug therapy , Morphine/administration & dosage , Pain/drug therapy , Animals , Bone Neoplasms/secondary , Disease Models, Animal , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Male , Pain/etiology , Prostatic Neoplasms/pathology , Rats , Rats, Wistar
7.
Pain Med ; 12(6): 923-41, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21539704

ABSTRACT

OBJECTIVE: Leconotide (CVID, AM336, CNSB004) is an omega conopeptide similar to ziconotide, which blocks voltage sensitive calcium channels. However, unlike ziconotide, which must be administered intrathecally, leconotide can be given intravenously because it is less toxic. This study investigated the antihyperalgesic potency of leconotide given intravenously alone and in combinations with morphine-administered intraperitoneally, in a rat model of bone cancer pain. DESIGN: Syngeneic rat prostate cancer cells AT3B-1 were injected into one tibia of male Wistar rats. The tumor expanded within the bone causing hyperalgesia to heat applied to the ipsilateral hind paw. Measurements were made of the maximum dose (MD) of morphine and leconotide given alone and in combinations that caused no effect in an open-field activity monitor, rotarod, and blood pressure and heart rate measurements. Paw withdrawal thresholds from noxious heat were measured. Dose response curves for morphine (0.312-5.0 mg/kg intraperitoneal) and leconotide (0.002-200 µg/kg intravenous) given alone were plotted and responses compared with those caused by morphine and leconotide in combinations. RESULTS: Leconotide caused minimal antihyperalgesic effects when administered alone. Morphine given alone intraperitoneally caused dose-related antihyperalgesic effects (ED(50) = 2.40 ± 1.24 mg/kg), which were increased by coadministration of leconotide 20 µg/kg (morphine ED(50) = 0.16 ± 1.30 mg/kg); 0.2 µg/kg (morphine ED(50) = 0.39 ± 1.27 mg/kg); and 0.02 µg/kg (morphine ED(50) = 1.24 ± 1.30 mg/kg). CONCLUSIONS: Leconotide caused a significant increase in reversal by morphine of the bone cancer-induced hyperalgesia without increasing the side effect profile of either drug. CLINICAL IMPLICATION: Translation into clinical practice of the method of analgesia described here will improve the quantity and quality of analgesia in patients with bone metastases. The use of an ordinary parenteral route for administration of the calcium channel blocker (leconotide) at low dose opens up the technique to large numbers of patients who could not have an intrathecal catheter for drug administration. Furthermore, the potentiating synergistic effect with morphine on hyperalgesia without increased side effects will lead to greater analgesia with improved quality of life.


Subject(s)
Analgesics , Bone Neoplasms/complications , Hyperalgesia/drug therapy , Morphine , Pain/drug therapy , Pain/etiology , omega-Conotoxins , Analgesics/administration & dosage , Analgesics/therapeutic use , Animals , Behavior, Animal , Bone Neoplasms/physiopathology , Dose-Response Relationship, Drug , Drug Synergism , Hyperalgesia/etiology , Injections, Intravenous , Male , Morphine/administration & dosage , Morphine/therapeutic use , Neoplasm Transplantation , Neuropsychological Tests , Pain Measurement , Rats , Rats, Wistar , Tibia/pathology , Tumor Cells, Cultured , omega-Conotoxins/administration & dosage , omega-Conotoxins/therapeutic use
8.
Pain Med ; 11(1): 106-18, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20447294

ABSTRACT

OBJECTIVE: This study determined the antihyperalgesic effect of CNSB002, a sodium channel blocker with antioxidant properties given alone and in combinations with morphine in rat models of inflammatory and neuropathic pain. DESIGN: Dose response curves for nonsedating doses of morphine and CNSB002 given intraperitoneally alone and together in combinations were constructed for antihyperalgesic effect using paw withdrawal from noxious heat in two rat pain models: carrageenan-induced paw inflammation and streptozotocin (STZ)-induced diabetic neuropathy. RESULTS: The maximum nonsedating doses were: morphine, 3.2 mg/kg; CNSB002 10.0 mg/kg; 5.0 mg/kg CNSB002 with morphine 3.2 mg/kg in combination. The doses calculated to cause 50% reversal of hyperalgesia (ED50) were 7.54 (1.81) and 4.83 (1.54) in the carrageenan model and 44.18 (1.37) and 9.14 (1.24) in the STZ-induced neuropathy model for CNSB002 and morphine, respectively (mg/kg; mean, SEM). These values were greater than the maximum nonsedating doses. The ED50 values for morphine when given in combination with CNSB002 (5 mg/kg) were less than the maximum nonsedating dose: 0.56 (1.55) in the carrageenan model and 1.37 (1.23) in the neuropathy model (mg/kg; mean, SEM). The antinociception after morphine (3.2 mg/kg) was increased by co-administration with CNSB002 from 28.0 and 31.7% to 114.6 and 56.9% reversal of hyperalgesia in the inflammatory and neuropathic models, respectively (P < 0.01; one-way analysis of variance-significantly greater than either drug given alone). CONCLUSIONS: The maximum antihyperalgesic effect achievable with nonsedating doses of morphine may be increased significantly when the drug is used in combination with CNSB002.


Subject(s)
Analgesics, Opioid/therapeutic use , Inflammation/drug therapy , Morphine/therapeutic use , Pain/drug therapy , Peripheral Nervous System Diseases/drug therapy , Piperazines/therapeutic use , Sodium Channel Blockers/therapeutic use , Amines/therapeutic use , Analgesics, Opioid/administration & dosage , Animals , Carrageenan , Cyclohexanecarboxylic Acids/therapeutic use , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/drug therapy , Dose-Response Relationship, Drug , Drug Synergism , GABA Modulators/therapeutic use , Gabapentin , Inflammation/chemically induced , Inflammation/complications , Male , Morphine/administration & dosage , Motor Activity/drug effects , Pain/etiology , Pain Measurement , Peripheral Nervous System Diseases/complications , Rats , Rats, Wistar , gamma-Aminobutyric Acid/therapeutic use
9.
Pain Med ; 11(2): 262-73, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20002322

ABSTRACT

OBJECTIVE: Leconotide is an omega-conotoxin that blocks neuronal voltage sensitive calcium channels. This study compared the antihyperalgesic potencies of leconotide and ziconotide given intravenously alone and in combinations with a potassium channel modulator flupirtine, given intraperitoneally, in a rat model of diabetic neuropathic pain. DESIGN: Rats were given streptozotocin (150 mg/kg ip) to induce diabetic neuropathy and hyperalgesia. Experiments were performed on diabetic rats with >or=30% hyperalgesia to noxious heat. Rats were given each conopeptide alone and with flupirtine. Open field activity monitoring and non-invasive blood pressure measurements were used to define the maximum doses and combinations that caused no side effects. Doses in a range up to maximum no side effect doses were tested for antihyperalgesic effects in rats with hyperalgesia. RESULTS: The maximum no side effect dose of leconotide (2 mg/kg intravenously) caused 51.7% reversal of hyperalgesia compared with 0.4% for the highest no side effect dose of ziconotide (0.02 mg/kg; P < 0.001, one-way anova). Leconotide caused dose-related antihyperalgesic effects that were potentiated by coadministration with flupirtine at doses that were ineffective when given alone. Leconotide (0.02 mg/kg) and flupirtine (5 mg/kg) caused 25.3 +/- 7.6 and -6 +/- 9.5% reversal of hyperalgesia, respectively when given alone but in combination they caused 84.1 +/- 7.2% reversal of hyperalgesia (P < 0.01; one-way anova). No such interaction occurred with ziconotide. CONCLUSION: Leconotide could have wider clinical applications than ziconotide. Unlike ziconotide, powerful antihyperalgesia without side effects can be achieved by intravenous administration of leconotide thus avoiding the need for an intrathecal injection.


Subject(s)
Analgesics, Non-Narcotic/therapeutic use , Calcium Channel Blockers/therapeutic use , Diabetic Neuropathies/drug therapy , Hyperalgesia/drug therapy , omega-Conotoxins/therapeutic use , Aminopyridines/therapeutic use , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/adverse effects , Animals , Blood Pressure/drug effects , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/adverse effects , Diabetes Mellitus, Experimental/complications , Diabetic Neuropathies/complications , Dose-Response Relationship, Drug , Drug Interactions , Hemodynamics/drug effects , Hot Temperature , Hyperalgesia/etiology , Injections, Intravenous , Male , Motor Activity/drug effects , Pain Measurement/drug effects , Rats , Rats, Wistar , omega-Conotoxins/administration & dosage , omega-Conotoxins/adverse effects
10.
Pain Med ; 10(5): 890-901, 2009.
Article in English | MEDLINE | ID: mdl-19682273

ABSTRACT

OBJECTIVE: Alphadolone is a neuroactive steroid that causes antinociception in rats and analgesia in humans by interaction with spinal cord GABA(A) receptors. This study investigated whether alphadolone could affect morphine tolerance. METHODS: Morphine tolerance was induced in rats with subcutaneous sustained release morphine emulsion (M-SR; 125 mg/kg/day). Tolerance was assessed by a blinded observer using tail flick latency (TFL) response to intraperitoneal (ip) injection of immediate release morphine (M-IR 6.25 mg/kg). Fifty-five rats given M-SR were divided into three groups: group A received 1.0 mL subcutaneous emulsion containing vehicle; groups B and C had emulsions injected subcutaneously at the same time as the M-SR (B-250 mg/kg alphadolone; C-alphaxalone 80 mg/kg). RESULTS: Tail flick latency responses (percentage of maximum possible effect [% MPE]) to M-IR were reduced from 89.6 +/- 2.5 pretreatment to 20.3 +/- 4.8 after M-SR treatment (mean +/- SEM; P < 0.001, one-way anova). Coadministration of alphadolone emulsion with the M-SR caused no sedation and prevented the occurrence of morphine tolerance. TFL responses to M-IR (6.25 mg/kg) given to morphine tolerant rats were 29 +/- 8% MPE whereas the TFL was 78.6 +/- 9.8% MPE when immediate release alphadolone (10 mg/kg ip) was injected at the same time as M-IR to tolerant rats (P < 0.001 one-way anova). Alphaxalone treatment caused sedation and no effects on morphine tolerance. CONCLUSIONS: We conclude that the alphadolone can prevent morphine tolerance and it also restores normal morphine antinociception in rats with established morphine tolerance. The lack of sedation suggests clinical utility in human pain states requiring morphine.


Subject(s)
Analgesics, Opioid/pharmacology , Analgesics/pharmacology , Morphine/pharmacology , Pregnanediones/pharmacology , Analgesics, Opioid/administration & dosage , Animals , Delayed-Action Preparations , Drug Tolerance , Emulsions , Hypnotics and Sedatives , Male , Morphine/administration & dosage , Pain Measurement/drug effects , Rats , Rats, Wistar
11.
Pain Med ; 9(7): 928-38, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18950446

ABSTRACT

OBJECTIVES: Flupirtine is an established clinical analgesic for mild to moderate musculoskeletal pain states. It has recently been shown to be a KCNQ2-3 potassium channel opener. These experiments were performed to see if this property could be useful in treating pain states characterized by central sensitization with the drug either given alone or in combination with morphine. DESIGN: Experiments were performed in rats in an observer-blinded fashion with vehicle controls. Nonsedating doses of flupirtine, morphine, and combinations containing both drugs were defined using the rotarod test and open-field activity monitoring. Dose-response relationships were determined for nonsedating doses of both drugs given alone and together in combination in causing antinociception in two nociception paradigms: carrageenan paw inflammation and streptozotocin-induced diabetic neuropathy. RESULTS: Flupirtine and morphine, when given alone at the highest nonsedating doses, caused slight to moderate antinociception in both paradigms. Flupirtine also caused significant increases in morphine antinociception in both models. In carrageenan paw inflammation, complete reversal of carrageenan-induced hyperalgesia was caused by 10 mg/kg flupirtine in combination with 0.4 mg/kg morphine. These doses of the two drugs were ineffective when given alone, but the combination caused complete antinociception in this model of inflammatory pain. In the diabetic neuropathy model, morphine 3.2 mg/kg given alone caused no significant antinociception. However, a lower dose of morphine (1.6 mg/kg shown to be ineffective when it was given alone) given in combination with flupirtine 10 mg/kg caused highly significant antinociceptive effects causing complete reversal of hyperalgesia caused by diabetic neuropathy (P < 0.001, one-way analysis of variance). This combination of drugs was not sedating. CONCLUSIONS: Flupirtine increases morphine antinociception without causing an increase in sedation. Flupirtine should be investigated as an adjunct analgesic with opioids for the management of patients with pain states involving central sensitization.


Subject(s)
Aminopyridines/administration & dosage , Disease Models, Animal , Morphine/administration & dosage , Neuralgia/diagnosis , Neuralgia/drug therapy , Pain Measurement/drug effects , Analgesics/administration & dosage , Animals , Dose-Response Relationship, Drug , Drug Therapy, Combination , Humans , Male , Rats , Rats, Wistar , Treatment Outcome
12.
Pain Med ; 9(7): 939-49, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18950447

ABSTRACT

OBJECTIVES: This study is a case series that was designed to provide data on the efficacy and the incidence and duration of adverse effects of flupirtine in the treatment of cancer-related neuropathic pain. DESIGN: This was an 8-day, open-label study of palliative care patients with neuropathic pain despite maximal opioid treatment. They received an initial dose of flupirtine 100 mg orally four times daily (QID) that could be titrated. Efficacy measures included: a neuropathic pain discriminant score; scales measuring average pain and quality of life activities; and a score of percentage pain relief. RESULTS: Ten patients were recruited. Only one patient was withdrawn because of side effects. Several pain measurements were used. All patients were able to participate in these measurements apart from two who did not understand the concept of percentage pain relief. There were significant reductions of average pain (P < 0.01) and neuropathic pain discriminant scores (P < 0.01), as well as an increase in percentage pain relief (P < 0.01). There was no statistically significant change in overall opioid use but 8/10 patients had some reduction in opioid use and three of those required substantially reduced doses of opioid when flupirtine was added to their treatment regime. Eight patients elected to continue to take flupirtine after the trial, two taking 200 mg QID and the others 100 mg QID. Of these eight, six said that flupirtine was of considerable help and two said it helped a little. All of these continued to take flupirtine with very good pain control until death, one of which was 18 months after the trial course. CONCLUSIONS: These results in humans follow animal studies that suggest a role for flupirtine in the treatment of neuropathic pain. This short duration open-label study in 10 subjects suggests that flupirtine may be useful in the treatment of neuropathic pain when used in combination with opioids.


Subject(s)
Aminopyridines/administration & dosage , Morphine/administration & dosage , Neuralgia/diagnosis , Neuralgia/drug therapy , Pain Measurement/drug effects , Adult , Aged , Aged, 80 and over , Analgesics/administration & dosage , Dose-Response Relationship, Drug , Drug Therapy, Combination , Humans , Male , Middle Aged , Palliative Care , Treatment Outcome
14.
BMC Anesthesiol ; 5(1): 2, 2005 Apr 02.
Article in English | MEDLINE | ID: mdl-15804361

ABSTRACT

BACKGROUND: Despite preclinical evidence suggesting a synergistic interaction between ketamine and opioids promoting analgesia, several clinical trials have not identified dosing regimens capable of eliciting a benefit in the co-administration of ketamine with opioids. METHODS: Ten healthy volunteers participated in a double blinded, randomised, placebo controlled, crossover laboratory study in order to determine whether a low dose of ketamine potentiated the antinociceptive effect of fentanyl without causing an increase in sedative effects. A battery of tests was used to assess both nociception and sedation including electrical current, pressure, thermal stimuli, psychometric tests, and both subjective and objective scores of sedation. Target controlled infusions of the study drugs were used. Ketamine and fentanyl were administered alone and in combination in a double-blinded randomised crossover design. Saline was used as the control, and propofol was used to validate the tests of sedation. Cardiovascular and respiratory parameters were also assessed. RESULTS: The electrical current pain threshold dose response curve of fentanyl combined with ketamine was markedly steeper than the dose response curve of fentanyl alone. While a ketamine serum concentration of 30 ng/ml did not result in a change in electrical pain threshold when administered alone, when it was added to fentanyl, the combination resulted in greater increase in pain threshold than that of fentanyl administered alone. When nociception was assessed using heat and pressure stimuli, ketamine did not potentiate the anti-nociceptive effect of fentanyl. There was no difference between the sedative effect of fentanyl and fentanyl in combination with ketamine as assessed by both subjective and objective measures of sedation. Cardiovascular and respiratory parameters were unaffected by the study drugs at the doses given. CONCLUSION: A serum concentration of ketamine that did not alter indices of sedation potentiated the antinociceptive effect of fentanyl. This potentiation of antinociception occurred without an increase in sedation suggesting that low steady doses of ketamine (30-120 ng/ml) might be combined with mu opioid agonists to improve their analgesic effect in a clinical setting. (296 words).

16.
Anesth Analg ; 98(6): 1512-1520, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15155299

ABSTRACT

UNLABELLED: Despite conflicting evidence regarding the safety of intrathecal midazolam from animal investigations, its clinical use is increasing. We investigated the potential of intrathecal midazolam to produce symptomatology suggestive of neurological damage. This study compared two cohorts of patients who received intrathecal anesthesia with or without intrathecal midazolam (2 mg). Eighteen risk factors were evaluated with respect to symptoms representing potential neurological complications. The definitions of these symptoms were made wide to maximize the chance of counting patients with neurological sequelae after intrathecal injections. Eleven-hundred patients were followed up prospectively during the first postoperative week by a hospital chart review and 1 mo later by a mailed questionnaire. Symptoms suggestive of neurological impairment, including motor or sensory changes and bladder or bowel dysfunction, were investigated. Intrathecal midazolam was not associated with an increased risk of neurologic symptoms. In contrast, neurologic symptoms were found to be increased by age >70 yr (relative risk, 8.72) and the occurrence of a blood-stained spinal tap (relative risk, 8.07). The administration of intrathecal midazolam, 2 mg, did not increase the occurrence of neurologic or urologic symptoms, as suggested by some preclinical animal experimentation. IMPLICATIONS: Intrathecal midazolam provides segmental analgesia, but conflicting animal studies have cast doubts on its safety. This investigation studied the effect of intrathecal midazolam by observing two cohorts of patients. In clinical practice, intrathecal midazolam (2 mg) did not increase adverse neurological symptoms compared with conventional therapies.


Subject(s)
Midazolam/administration & dosage , Midazolam/adverse effects , Surveys and Questionnaires , Aged , Chi-Square Distribution , Cohort Studies , Confidence Intervals , Female , Follow-Up Studies , Humans , Injections, Spinal , Logistic Models , Male , Middle Aged , Nervous System Diseases/chemically induced , Risk Factors , Statistics, Nonparametric
17.
Anesth Analg ; 98(6): 1521-1527, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15155300

ABSTRACT

UNLABELLED: Recent investigations have sought to improve intrathecal analgesia by combining opioids with other classes of analgesics. In this study we assessed the ability of intrathecal midazolam to increase the potency and duration of the analgesic effects of intrathecal fentanyl without causing adverse effects. Thirty parturients with cervical dilations 2-6 cm were randomized to receive either intrathecal midazolam 2 mg, fentanyl 10 micro g, or both combined to initiate analgesia. Pain scores were recorded before and at 5-min intervals for 30 min after the injection and then every 30 minutes until the patient requested further analgesia. The presence and severity of nausea, emesis, pruritus, headache, and sedation, in addition to arterial blood pressure, heart rate, respiratory rate, sensory changes to ice, motor impairment, cardiotocograph, and Apgar score were also recorded. The parturients were assessed after 2 days and 1 mo for neurologic impairment. Preinjection pain scores were unaltered by intrathecal midazolam alone and moderately decreased by fentanyl. Intrathecal midazolam increased the analgesic effect of fentanyl. No treatment altered cardiorespiratory variables or caused motor impairment. The addition of intrathecal midazolam to fentanyl did not increase the occurrence of any maternal adverse event or abnormalities on the cardiotocograph. We conclude that intrathecal midazolam enhanced the analgesic effect of fentanyl without increasing maternal or fetal adverse effects. IMPLICATIONS: Treatment of labor pain with epidural injections of local anesthetic is complicated by decreases in arterial blood pressure and leg weakness. This study showed that a mixture of two drugs, fentanyl and midazolam, could provide powerful pain relief when the drugs were given together spinally without such side effects.


Subject(s)
Fentanyl/administration & dosage , Labor, Obstetric/drug effects , Midazolam/administration & dosage , Pain/drug therapy , Adult , Analysis of Variance , Chi-Square Distribution , Drug Therapy, Combination , Female , Humans , Injections, Spinal , Pain/physiopathology , Pregnancy , Statistics, Nonparametric , Time Factors
18.
Anesth Analg ; 98(6): 1557-1565, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15155305

ABSTRACT

UNLABELLED: Clinicians need a simple, fast, reliable, and inexpensive way of identifying the evidence base relevant to their clinical practice. It is often believed that the only way to identify all relevant evidence is to perform hand-searches of the literature to supplement computer searches; this is complex and labor intensive. However, most of quality randomized controlled trials cited in systematic reviews in pain medicine are listed in computer databases. We performed two studies to investigate the efficiency-in terms of sensitivity, specificity, and precision-of three computer search strategies: Optimally Sensitive Search Strategy, which is used by the Cochrane Collaboration; RCT.pt, a standard MEDLINE strategy; and DBRCT.af, which is a new single-line computer algorithm based on the assumption that double-blinded, randomized controlled trials would be indexed with "double-blind," "random," or variations of these terms in MEDLINE and EMBASE. DBRCT.af was found to be highly sensitive (97%) in identifying quality randomized controlled trials in pain medicine. The precision (ratio of randomized controlled trials to the number of nonrandomized trials identified) was 82%, and the specificity in excluding the nonrandomized controlled trials was 98%. We conclude that clinicians can now use DBRCT.af to update and conduct de novo systematic reviews in pain-relief research. IMPLICATIONS: Quality evidence about what is good clinical practice in pain treatment is buried in the medical literature among large quantities of other information. This article describes how any clinician with access to the Internet can identify those quality studies reliably, quickly, and inexpensively.


Subject(s)
Analgesia/statistics & numerical data , Evidence-Based Medicine/methods , Information Storage and Retrieval/methods , Pain Management , Randomized Controlled Trials as Topic/statistics & numerical data , Evidence-Based Medicine/statistics & numerical data , Humans , Information Storage and Retrieval/statistics & numerical data , Internet/statistics & numerical data , Pain Measurement/methods , Pain Measurement/statistics & numerical data
19.
BMC Pharmacol ; 2: 20, 2002 Nov 19.
Article in English | MEDLINE | ID: mdl-12441008

ABSTRACT

BACKGROUND: Several studies have shown that muscarinic cholinergic agonists cause antinociception in humans and animals when given by both spinal and non-spinal parenteral routes. It is uncertain which subtype of muscarinic receptor is involved in spinally mediated antinociceptive effects caused by these drugs. The cholinergic receptor agonists McN-A-343 (M1 selective; 3.89 to 389 nmol) and carbachol (non-selective; 0.029 to 29 nmol) were used in a rat acute pain model to investigate the involvement of M1 and non-M1 subtypes in spinally mediated antinociception. The drugs were injected intrathecally and results from experiments in which drug actions were carefully confined to the spinal cord were used to construct agonist dose response curves. RESULTS: McN-A-343 frequently diffused rostrally to the brain, away from the lumbosacral site of injection. Thus, in spite of its receptor subtype selectivity, McN-A-343 is a poor probe to use in attempting to identify receptor subtypes involved in spinal cord antinociceptive systems. However, in some experiments McN-A-343 caused spinally mediated antinociception assessed by the electrical current threshold test. Antinociception assessed by the tail flick latency test with intrathecal McN-A-343 was observed and found to involve supraspinal mechanisms. Carbachol caused spinally mediated antinociception assessed by both electrical current threshold and tail flick latency. CONCLUSIONS: The results suggest that M1 receptors are involved in spinally mediated antinociception revealed by electrical current threshold; other cholinergic receptors (non-M1) are involved in thermal antinociception at the spinal cord. This contrasts with previous work on spinally mediated cholinergic antinociception. These differences are believed to be due to difficulties in restricting the action of these drugs to the spinal cord.


Subject(s)
(4-(m-Chlorophenylcarbamoyloxy)-2-butynyl)trimethylammonium Chloride/therapeutic use , Cholinergic Agents/therapeutic use , Pain/drug therapy , Spinal Cord/drug effects , (4-(m-Chlorophenylcarbamoyloxy)-2-butynyl)trimethylammonium Chloride/pharmacology , Animals , Carbachol/therapeutic use , Cholinergic Agents/pharmacology , Disease Models, Animal , Male , Muscarinic Agonists/therapeutic use , Pain Measurement/drug effects , Rats , Rats, Wistar , Receptor, Muscarinic M1/metabolism
20.
Pain ; 48(1): 5-12, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1531383

ABSTRACT

This paper describes a prospective, double blind, randomised and dummy-controlled trial in 28 patients with chronic mechanical low back pain presenting to the York Pain Clinic. The therapeutic effects of epidural methyl prednisolone (80 mg) were compared with intrathecal midazolam (2 mg). All the patients had pain for a considerable length of time (range: 1-35 years) and all had received previous treatments which had failed. The two groups of patients were comparable in terms of pain duration, demography, extent of disability, anxiety and depression and pain locus of control. The pain was assessed before and for 2 months after treatment using the short form McGill Pain Questionnaire as well as visual analogue and verbal rating scales for sensory and affective components of their pain experience; patients also completed a pain diary. Both treatments caused a similar improvement in one-half to three-quarters of the patients for 2 months in patterns of activity and sleep as well as in the sensory and affective components of the pain. However, although the improvement in the two groups was similar, all the patients treated with steroid were either taking more or the same amount of self-administered analgesic medication after their treatment, whereas between one-third and one-half of the midazolam-treated patients took less medication during the 2 month follow-up period. We conclude that intrathecal midazolam is an effective treatment for chronic mechanical low back pain. The mechanism responsible for this effect is discussed.


Subject(s)
Back Pain/drug therapy , Methylprednisolone/therapeutic use , Midazolam/therapeutic use , Adult , Aged , Back Pain/physiopathology , Back Pain/psychology , Double-Blind Method , Female , Humans , Injections, Epidural , Injections, Spinal , Male , Methylprednisolone/administration & dosage , Midazolam/administration & dosage , Middle Aged , Pain Measurement , Pilot Projects , Prospective Studies , Surveys and Questionnaires , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...