Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Neurochem Res ; 45(6): 1256-1267, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31749072

ABSTRACT

In tribute to our friend and colleague Michael Robinson, we review his involvement in the identification, characterization and localization of the metallopeptidase glutamate carboxypeptidase II (GCPII), originally called NAALADase. While Mike was characterizing NAALADase in the brain, the protein was independently identified by other laboratories in human prostate where it was termed prostate specific membrane antigen (PSMA) and in the intestines where it was named Folate Hydrolase 1 (FOLH1). It was almost a decade to establish that NAALADase, PSMA, and FOLH1 are encoded by the same gene. The enzyme has emerged as a therapeutic target outside of the brain, with the most notable progress made in the treatment of prostate cancer and inflammatory bowel disease (IBD). PSMA-PET imaging with high affinity ligands is proving useful for the clinical diagnosis and staging of prostate cancer. A molecular radiotherapy based on similar ligands is in trials for metastatic castration-resistant prostate cancer. New PSMA inhibitor prodrugs that preferentially block kidney and salivary gland versus prostate tumor enzyme may improve the clinical safety of this radiotherapy. The wide clinical use of PSMA-PET imaging in prostate cancer has coincidentally led to clinical documentation of GCPII upregulation in a wide variety of tumors and inflammatory diseases, likely associated with angiogenesis. In IBD, expression of the FOLH1 gene that codes for GCPII is strongly upregulated, as is the enzymatic activity in diseased patient biopsies. In animal models of IBD, GCPII inhibitors show substantial efficacy, suggesting potential theranostic use of GCPII ligands for IBD.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain/drug effects , Brain/metabolism , Glutamate Carboxypeptidase II/antagonists & inhibitors , Glutamate Carboxypeptidase II/metabolism , Prodrugs/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Humans , Male , Prodrugs/chemistry , Prodrugs/pharmacology , Prostate/drug effects , Prostate/metabolism
2.
Eur J Pharmacol ; 840: 89-103, 2018 Dec 05.
Article in English | MEDLINE | ID: mdl-30268665

ABSTRACT

Chemotherapy-Induced Peripheral Neurotoxicity (CIPN) is often dose-limiting and impacts life quality and survival of cancer patients. Ghrelin agonists have neuroprotectant effects and may have a role in treating or preventing CIPN. We evaluated the CNS-penetrant ghrelin agonist HM01 in three experimental models of CIPN at doses of 3-30 mg/kg p.o. daily monitoring orexigenic properties, nerve conduction, mechanical allodynia, and intra-epidermal nerve fiber density (IENFD). In a cisplatin-based study, rats were dosed daily for 3 days (0.5 mg/kg i.p.) + HM01. Cisplatin treatment induced mechanical hypersensitivity which was significantly reduced by HM01. In a second study, oxaliplatin was administered to mice (6 mg/kg i.p. 3 times/week for 4 weeks) resulting in significant digital nerve conduction velocity (NCV) deficits and reduction of IENFD. Concurrent HM01 dose dependently prevented the decline in NCV and attenuated the reduction in IENFD. Pharmacokinetic studies showed HM01 accumulation in the dorsal root ganglia and sciatic nerves which reached concentrations > 10 fold that of plasma. In a third model, HM01 was tested in preventive and therapeutic paradigms in a bortezomib-based rat model (0.2 mg/kg i.v., 3 times/week for 8 weeks). In the preventive setting, HM01 blocked bortezomib-induced hyperalgesia and IENFD reduction at all doses tested. In the therapeutic setting, significant effect was observed, but only at the highest dose. Altogether, the robust peripheral nervous system penetration of HM01 and its ability to improve multiple oxaliplatin-, cisplatin-, and bortezomib-induced neurotoxicities suggest that HM01 may be a useful neuroprotective adjuvant for CIPN.


Subject(s)
Antineoplastic Agents/adverse effects , Benzene Derivatives/pharmacology , Ghrelin/agonists , Nervous System/drug effects , Neuroprotective Agents/pharmacology , Animals , Body Weight/drug effects , Cisplatin/adverse effects , Dose-Response Relationship, Drug , Eating/drug effects , Female , Male , Mice , Neural Conduction/drug effects , Piperidines , Rats
3.
Mol Cancer Ther ; 17(9): 1824-1832, 2018 09.
Article in English | MEDLINE | ID: mdl-30181331

ABSTRACT

The broadly active glutamine antagonist 6-diazo-5-oxo-L-norleucine (DON) has been studied for 60 years as a potential anticancer therapeutic. Clinical studies of DON in the 1950s using low daily doses suggested antitumor activity, but later phase I and II trials of DON given intermittently at high doses were hampered by dose-limiting nausea and vomiting. Further clinical development of DON was abandoned. Recently, the recognition that multiple tumor types are glutamine-dependent has renewed interest in metabolic inhibitors such as DON. Here, we describe the prior experience with DON in humans. Evaluation of past studies suggests that the major impediments to successful clinical use included unacceptable gastrointestinal (GI) toxicities, inappropriate dosing schedules for a metabolic inhibitor, and lack of targeted patient selection. To circumvent GI toxicity, prodrug strategies for DON have been developed to enhance delivery of active compound to tumor tissues, including the CNS. When these prodrugs are administered in a low daily dosing regimen, appropriate for metabolic inhibition, they are robustly effective without significant toxicity. Patients whose tumors have genetic, metabolic, or imaging biomarker evidence of glutamine dependence should be prioritized as candidates for future clinical evaluations of novel DON prodrugs, given either as monotherapy or in rationally directed pharmacologic combinations. Mol Cancer Ther; 17(9); 1824-32. ©2018 AACR.


Subject(s)
Diazooxonorleucine/therapeutic use , Glutamine/antagonists & inhibitors , Neoplasms/drug therapy , Prodrugs/therapeutic use , Animals , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/therapeutic use , Diazooxonorleucine/adverse effects , Diazooxonorleucine/chemistry , Glutamine/metabolism , Humans , Molecular Structure , Nausea/chemically induced , Neoplasms/metabolism , Prodrugs/adverse effects , Prodrugs/chemistry , Vomiting/chemically induced
4.
Cancer Res ; 78(3): 817-829, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29191802

ABSTRACT

Chemotherapy-induced peripheral neuropathy (CIPN) is a major cause of disability in cancer survivors. CIPN investigations in preclinical model systems have focused on either behaviors or acute changes in nerve conduction velocity (NCV) and amplitude, but greater understanding of the underlying nature of axonal injury and its long-term processes is needed as cancer patients live longer. In this study, we used multiple independent endpoints to systematically characterize CIPN recovery in mice exposed to the antitubulin cancer drugs eribulin, ixabepilone, paclitaxel, or vinorelbine at MTDs. All of the drugs ablated intraepidermal nerve fibers and produced axonopathy, with a secondary disruption in myelin structure within 2 weeks of drug administration. In addition, all of the drugs reduced sensory NCV and amplitude, with greater deficits after paclitaxel and lesser deficits after ixabepilone. These effects correlated with degeneration in dorsal root ganglia (DRG) and sciatic nerve and abundance of Schwann cells. Although most injuries were fully reversible after 3-6 months after administration of eribulin, vinorelbine, and ixabepilone, we observed delayed recovery after paclitaxel that produced a more severe, pervasive, and prolonged neurotoxicity. Compared with other agents, paclitaxel also displayed a unique prolonged exposure in sciatic nerve and DRG. The most sensitive indicator of toxicity was axonopathy and secondary myelin changes accompanied by a reduction in intraepidermal nerve fiber density. Taken together, our findings suggest that intraepidermal nerve fiber density and changes in NCV and amplitude might provide measures of axonal injury to guide clinical practice.Significance: This detailed preclinical study of the long-term effects of widely used antitubulin cancer drugs on the peripheral nervous system may help guide clinical evaluations to improve personalized care in limiting neurotoxicity in cancer survivors. Cancer Res; 78(3); 817-29. ©2017 AACR.


Subject(s)
Ganglia, Spinal/drug effects , Microtubules/drug effects , Peripheral Nervous System Diseases/chemically induced , Recovery of Function/drug effects , Schwann Cells/drug effects , Sciatic Nerve/drug effects , Tubulin Modulators/toxicity , Acute Disease , Animals , Cells, Cultured , Female , Ganglia, Spinal/injuries , Ganglia, Spinal/pathology , Mice , Mice, Inbred BALB C , Microtubules/pathology , Peripheral Nervous System Diseases/pathology , Schwann Cells/pathology , Sciatic Nerve/injuries , Sciatic Nerve/pathology
5.
JCI Insight ; 1(12)2016 Aug 04.
Article in English | MEDLINE | ID: mdl-27536732

ABSTRACT

Recent gene-profiling analyses showed significant upregulation of the folate hydrolase (FOLH1) gene in the affected intestinal mucosa of patients with inflammatory bowel disease (IBD). The FOLH1 gene encodes a type II transmembrane glycoprotein termed glutamate carboxypeptidase II (GCPII). To establish that the previously reported increased gene expression was functional, we quantified the glutamate carboxypeptidase enzymatic activity in 31 surgical specimens and report a robust 2.8- to 41-fold increase in enzymatic activity in the affected intestinal mucosa of IBD patients compared with an uninvolved area in the same patients or intestinal mucosa from healthy controls. Using a human-to-mouse approach, we next showed a similar enzymatic increase in two well-validated IBD murine models and evaluated the therapeutic effect of the potent FOLH1/ GCPII inhibitor 2-phosphonomethyl pentanedioic acid (2-PMPA) (IC50 = 300 pM). In the dextran sodium sulfate (DSS) colitis model, 2-PMPA inhibited the GCPII activity in the colonic mucosa by over 90% and substantially reduced the disease activity. The significance of the target was confirmed in FOLH1-/- mice who exhibited resistance to DSS treatment. In the murine IL-10-/- model of spontaneous colitis, daily 2-PMPA treatment also significantly reduced both macroscopic and microscopic disease severity. These results provide the first evidence of FOLH1/GCPII enzymatic inhibition as a therapeutic option for IBD.

6.
Cancer Res ; 76(11): 3332-9, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27197173

ABSTRACT

Chemotherapy-induced peripheral neuropathy is a dose-limiting side effect of many antineoplastic agents, but the mechanisms underlying the toxicities are unclear. At their MTDs, the microtubule-binding drugs paclitaxel and ixabepilone induce more severe neuropathy in mice relative to eribulin mesylate, paralleling their toxicity profiles in clinic. We hypothesized that the severity of their neurotoxic effects might be explained by the levels at which they accumulate in the peripheral nervous system. To test this hypothesis, we compared their pharmacokinetics and distribution in peripheral nerve tissue. After administration of a single intravenous dose, each drug was rapidly cleared from plasma but all persisted in the dorsal root ganglia (DRG) and sciatic nerve (SN) for up to 72 hours. Focusing on paclitaxel and eribulin, we performed a 2-week MTD-dosing regimen, followed by a determination of drug pharmacokinetics, tissue distribution, and multiple functional measures of peripheral nerve toxicity for 4 weeks. Consistent with the acute dosing study, both drugs persisted in peripheral nervous tissues for weeks, in contrast to their rapid clearance from plasma. Notably, although eribulin exhibited greater DRG and SN penetration than paclitaxel, the neurotoxicity observed functionally was consistently more severe with paclitaxel. Overall, our results argue that sustained exposure of microtubule-binding chemotherapeutic agents in peripheral nerve tissues cannot by itself account for their associated neurotoxicity. Cancer Res; 76(11); 3332-9. ©2016 AACR.


Subject(s)
Apoptosis/drug effects , Ganglia, Spinal/pathology , Microtubules/pathology , Paclitaxel/pharmacology , Peripheral Nervous System Diseases/pathology , Sciatic Nerve/pathology , Animals , Blotting, Western , Cell Proliferation , Dose-Response Relationship, Drug , Electrophysiology , Epothilones/pharmacokinetics , Epothilones/pharmacology , Female , Furans/pharmacokinetics , Furans/pharmacology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ketones/pharmacokinetics , Ketones/pharmacology , Mice , Mice, Inbred BALB C , Microtubules/drug effects , Microtubules/metabolism , Paclitaxel/pharmacokinetics , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/metabolism , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Time Factors , Tissue Distribution , Tubulin Modulators/pharmacokinetics , Tubulin Modulators/pharmacology
7.
J Transl Med ; 13: 170, 2015 May 29.
Article in English | MEDLINE | ID: mdl-26021605

ABSTRACT

BACKGROUND: Propofol is a safe and widely used intravenous anesthetic agent, for which additional clinical uses including treatment of migraine, nausea, pain and anxiety have been proposed (Vasileiou et al. Eur J Pharmacol 605:1-8, 2009). However, propofol suffers from several disadvantages as a therapeutic outside anesthesia including its limited aqueous solubility and negligible oral bioavailability. The purpose of the studies described here was to evaluate, in both animals and human volunteers, whether fospropofol (a water soluble phosphate ester prodrug of propofol) would provide higher propofol bioavailability through non-intravenous routes. METHODS: Fospropofol was administered via intravenous, oral and intraduodenal routes to rats. Pharmacokinetic and pharmacodynamic parameters were then evaluated. Based on the promising animal data we subsequently conducted an oral and intraduodenal pharmacokinetic/pharmacodynamic study in human volunteers. RESULTS: In rats, bioavailability of propofol from fospropofol delivered orally was found to be appreciable, in the order of around 20-70%, depending on dose. Availability was especially marked following fospropofol administration via the intraduodenal route, where bioavailability approximated 100%. Fospropofol itself was not appreciably bioavailable when administered by any route except for intravenous. Pharmacologic effect following oral fospropofol was confirmed by observation of sedation and alleviation of thermal hyperalgesia in the rat chronic constrictive injury model of neuropathic pain. The human data also showed systemic availability of propofol from fospropofol administration via oral routes, a hereto novel finding. Assessment of sedation in human volunteers was correlated with pharmacokinetic measurements. CONCLUSIONS: These data suggest potential utility of oral administration of fospropofol for various therapeutic indications previously considered for propofol.


Subject(s)
Gastrointestinal Tract/drug effects , Healthy Volunteers , Propofol/analogs & derivatives , Administration, Intravenous , Administration, Oral , Adolescent , Adult , Analgesics/pharmacology , Animals , Biological Availability , Dose-Response Relationship, Drug , Drug Administration Routes , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/pharmacokinetics , Hypnotics and Sedatives/pharmacology , Male , Middle Aged , Propofol/administration & dosage , Propofol/adverse effects , Propofol/pharmacokinetics , Rats, Sprague-Dawley , Young Adult
8.
Pharmacol Res Perspect ; 3(6): e00198, 2015 12.
Article in English | MEDLINE | ID: mdl-27022470

ABSTRACT

The preclinical pharmacodynamic and pharmacokinetic properties of 4-methylbenzyl (3S, 4R)-3-fluoro-4-[(Pyrimidin-2-ylamino) methyl] piperidine-1-carboxylate (CERC-301), an orally bioavailable selective N-methyl-D-aspartate (NMDA) receptor subunit 2B (GluN2B) antagonist, were characterized to develop a translational approach based on receptor occupancy (RO) to guide CERC-301 dose selection in clinical trials of major depressive disorder. CERC-301 demonstrated high-binding affinity (K i, 8.1 nmol L(-1)) specific to GluN2B with an IC 50 of 3.6 nmol L(-1) and no off-target activity. CERC-301 efficacy was demonstrated in the forced swim test with an efficacy dose (ED 50) of 0.3-0.7 mg kg(-1) (RO, 30-50%); increase in locomotor activity was observed at ED 50 of 2 mg kg(-1), corresponding to an RO of 75%. The predicted 50% RO concentration (Occ50) in humans was 400 nmol L(-1), similar to that predicted for rat, dog, and monkey (300, 200, and 400 nmol L(-1), respectively). Safety pharmacology and neurotoxicity studies raised no specific safety concerns. A first-in-human study in healthy males demonstrated a dose-proportional pharmacokinetic profile, with T max of ~1 h and t 1/2 of 12-17 h. Based on the preclinical and pharmacodynamic data, doses of ≥8 mg in humans are hypothesized to have an acceptable safety profile and result in clinically relevant peak plasma exposure.

9.
J Pharmacol Exp Ther ; 346(3): 406-13, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23776202

ABSTRACT

Glutamate carboxypeptidase II (GCP II) is a therapeutic target in neurologic disorders associated with excessive activation of glutamatergic systems. The potent, orally bioavailable GCP II inhibitor 2-(3-mercaptopropyl) pentanedioic acid (2-MPPA) is effective in preclinical models of diseases where excess glutamate release is implicated, including neuropathic pain, and was the first GCP II inhibitor to be administered to man. The relationships between dosing regimen, pharmacokinetics, and analgesia in a neuropathic pain model were examined in rats to aid development of clinical dosing. The efficacy of oral 2-MPPA in the chronic constrictive injury model was not simply related to plasma concentrations. Even though maximal concentrations were observed within 1 hour of dosing, the analgesic effect took at least 8 days of daily dosing to become significant. The delay was not due to tissue drug accumulation since inhibitory concentrations of the drug were achieved in the nerve within 1 hour of dosing. There was also no accumulation of drug in plasma or tissue after multiple daily dosing. Effects were dependent on reaching a threshold concentration since dividing the daily dose led to a loss of effect. The analgesic effect outlasted plasma exposure and was maintained for days even after daily dosing was halted. The delayed onset, dependence on threshold plasma concentration, and sustained effects after exposure support the hypothesis that an indirect, long-lived mechanism of action exists. Although these longer lasting secondary mechanisms are not yet identified, daily clinical dosing of a GCP II inhibitor seems justified.


Subject(s)
Glutamate Carboxypeptidase II/antagonists & inhibitors , Glutarates/pharmacokinetics , Glutarates/therapeutic use , Neuralgia/drug therapy , Sulfhydryl Compounds/pharmacokinetics , Sulfhydryl Compounds/therapeutic use , Animals , Area Under Curve , Biological Availability , Constriction, Pathologic/complications , Constriction, Pathologic/drug therapy , Dose-Response Relationship, Drug , Half-Life , Hot Temperature , Hyperalgesia/drug therapy , Male , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley
10.
J Pharmacol Exp Ther ; 343(3): 746-54, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22988061

ABSTRACT

Peripheral neuropathy from nerve trauma is a significant problem in the human population and often constitutes a dose-limiting toxicity in patients receiving chemotherapy. (3-2-Mercaptoethyl)biphenyl-2,3-dicarboxylic acid (E2072) is a potent (K(i) = 10 nM), selective, and orally available inhibitor of glutamate carboxypeptidase II (GCPII). Here, we report that E2072 attenuates hyperalgesia and nerve conduction velocity deficits in preclinical rodent models of neuropathic pain and oxaliplatin-induced neuropathy. In the chronic constrictive injury model, orally administered E2072 reversed pre-existing thermal hyperalgesia in rats in a dose-dependent fashion with a minimally effective dose of 0.1 mg/kg/day. It is noteworthy that multiple days of dosing of E2072 were required before analgesia was realized even though GCPII inhibitory exposures were achieved on the first day of dosing. In addition, analgesia was found to persist for up to 7 days after cessation of dosing, consistent with E2072's pharmacokinetic profile and sustained exposure. Furthermore, in a chronic oxaliplatin-induced neuropathy model (6 mg/kg i.p. oxaliplatin twice weekly for 4 weeks), female BALB/c mice receiving daily oral E2072 at 1.0 and 0.1 mg/kg displayed no deficits in either caudal or digital velocity compared with significant deficits observed in mice treated with oxaliplatin alone (12 ± 3 and 9 ± 2%, respectively). Similar findings were seen with oxaliplatin-induced digital and caudal amplitude deficits. It is noteworthy that E2072 showed no interference with the antineoplastic efficacy of oxaliplatin in mice bearing leukemia (L1210), even at doses 100 times its neuroprotective/analgesic dose, indicating a selective effect on neuropathy. These data support the therapeutic utility of GCPII inhibitors in neuropathy and neuropathic pain.


Subject(s)
Benzoates/therapeutic use , Enzyme Inhibitors/therapeutic use , Glutamate Carboxypeptidase II/antagonists & inhibitors , Hyperalgesia/drug therapy , Neural Conduction/drug effects , Neuralgia/drug therapy , Sciatic Neuropathy/drug therapy , Sulfhydryl Compounds/therapeutic use , Administration, Oral , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzoates/administration & dosage , Benzoates/chemistry , Benzoates/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Female , Hyperalgesia/enzymology , Hyperalgesia/physiopathology , Leukemia L1210/drug therapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Molecular Structure , Neuralgia/enzymology , Neuralgia/physiopathology , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Rats , Rats, Sprague-Dawley , Sciatic Neuropathy/enzymology , Sciatic Neuropathy/physiopathology , Sulfhydryl Compounds/administration & dosage , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/pharmacokinetics , Tissue Distribution
11.
Anesth Analg ; 100(3): 701-706, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15728055

ABSTRACT

GPI 15715 is the first water-soluble propofol prodrug that has been studied in humans. Present propofol lipid formulations have well known undesirable properties, for example, pain on injection and increased triglyceride concentrations. We investigated whether GPI 15715 is suitable to achieve and maintain moderate sedation for 2 h. Six male and six female volunteers received a target-controlled infusion of GPI 15715, with an initial propofol target concentration of 1.8 microg/mL and the possibility to adjust the propofol target once after 1 h. Propofol concentrations, the bispectral index, and modified Observer's Assessment of Alertness/Sedation Scale (MOAA/S) scores were monitored. The median MOAA/S score was 4 during the first hour and was 3 during the second hour of infusion. The propofol target had to be changed to 2.4 microg/mL in seven volunteers and to 3.0 microg/mL in two volunteers. A propofol concentration of 1.9 microg/mL had the highest probability to result in an MOAA/S score of 3, which corresponds with moderate sedation. We observed no serious side effects. We conclude that GPI 15715 produces excellent sedation.


Subject(s)
Hypnotics and Sedatives/administration & dosage , Propofol/analogs & derivatives , Propofol/administration & dosage , Adult , Blood Pressure/drug effects , Female , Heart Rate/drug effects , Humans , Male , Propofol/blood
12.
Anesthesiology ; 101(3): 626-39, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15329587

ABSTRACT

BACKGROUND: GPI 15715 is a new water-soluble prodrug that is hydrolyzed to release propofol. The objectives of this crossover study in volunteers were to investigate the pharmacokinetics and pharmacodynamics of GPI 15715 in comparison with propofol emulsion. METHODS: In two separate sessions, nine healthy male volunteers (19-35 yr, 70-86 kg) received GPI 15715 and propofol emulsion as a target controlled infusion over 60 min. In the first 20 min, the propofol target concentration increased linearly to 5 microg/ml. Subsequently, the targets were reduced to 3 microg/ml and 1.5 microg/ml for 20 min each. The plasma concentrations of GPI 15715 and propofol were measured from arterial and venous blood samples up to 24 h and pharmacokinetics were analyzed. The pharmacodynamic effect was measured by the median frequency of the power spectrum of the electroencephalogram, and a sigmoid model with effect compartment was fitted to the data. RESULTS: Compared with propofol emulsion, propofol from GPI 15715 showed a different disposition function and especially larger volumes of distribution. The propofol effect site concentration for half maximum effect was 2.0 +/- 0.5 microg/ml for GPI 15715 and 3.0 +/- 0.7 microg/ml for propofol emulsion (P < 0.05). Propofol from GPI 15715 did not show a hysteresis between plasma concentration and effect. CONCLUSIONS: Compared with propofol emulsion, propofol from GPI 15715 showed different pharmacokinetics and pharmacodynamics, particularly a higher potency with respect to concentration. These differences may indicate an influence of the formulation.


Subject(s)
Anesthetics, Intravenous/pharmacology , Anesthetics, Intravenous/pharmacokinetics , Prodrugs/pharmacology , Prodrugs/pharmacokinetics , Propofol/analogs & derivatives , Propofol/pharmacology , Propofol/pharmacokinetics , Adult , Algorithms , Anesthetics, Intravenous/administration & dosage , Chemistry, Pharmaceutical , Cross-Over Studies , Electroencephalography/drug effects , Hemodynamics/drug effects , Humans , Infusions, Intravenous , Male , Models, Statistical , Prodrugs/administration & dosage , Propofol/administration & dosage , Sleep/drug effects
13.
Anesthesiology ; 99(2): 303-13, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12883403

ABSTRACT

BACKGROUND: GPI 15715 (AQUAVAN injection) is a new water-soluble prodrug which is hydrolyzed to release propofol. The objectives of this first study in humans were to investigate the safety, tolerability, pharmacokinetics, and clinical pharmacodynamics of GPI 15715. METHODS: Three groups of three healthy male volunteers (aged 19-35 y, 67-102 kg) received 290, 580, and 1,160 mg GPI 15715 as a constant rate infusion over 10 min. The plasma concentrations of GPI 15715 and propofol were measured from arterial and venous blood samples up to 24 h. Pharmacokinetics were analyzed with compartment models. Pharmacodynamics were assessed by clinical signs. RESULTS: GPI 15715 was well tolerated without pain on injection. Two subjects reported a transient unpleasant sensation of burning or tingling at start of infusion. Loss of consciousness was achieved in none with 290 mg and in one subject with 580 mg. After 1,160 mg, all subjects experienced loss of consciousness at propofol concentrations of 2.1 +/- 0.6 microg/ml. A two-compartment model for GPI 15715 (central volume of distribution, 0.07 l/kg; clearance, 7 ml. kg-1 min-1; terminal half-life, 46 min) and a three-compartment model for propofol (half-lives: 2.2, 20, 477 min) best described the data. The maximum decrease of blood pressure was 25%; the heart rate increased by approximately 35%. There were no significant laboratory abnormalities. CONCLUSIONS: Compared with propofol lipid emulsion, the potency seemed to be higher with respect to plasma concentration but was apparently less with respect to dose. Pharmacokinetic simulations showed a longer time to peak propofol concentration after a bolus dose and a longer context-sensitive half-time.


Subject(s)
Anesthetics, Intravenous/pharmacology , Anesthetics, Intravenous/pharmacokinetics , Prodrugs/pharmacology , Prodrugs/pharmacokinetics , Propofol/pharmacology , Propofol/pharmacokinetics , Adult , Anesthesia, Intravenous , Anesthetics, Intravenous/adverse effects , Area Under Curve , Blood Pressure/drug effects , Consciousness/drug effects , Half-Life , Heart Rate/drug effects , Humans , Male , Models, Biological , Prodrugs/adverse effects , Propofol/adverse effects , Propofol/analogs & derivatives , Respiratory Mechanics/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...