S(+)-ketamine Effect on Experimental Pain and Cardiac Output: A Population Pharmacokinetic-Pharmacodynamic Modeling Study in Healthy Volunteers

Background:Low-dose ketamine behaves as an analgesic in the treatment of acute and chronic pain. To further understand ketamine’s therapeutic profile, the authors performed a population pharmacokinetic-pharmacodynamic analysis of the S(+)-ketamine analgesic and nonanalgesic effects in healthy volunteers. Methods:Ten men and ten women received a 2-h S(+)-ketamine infusion. The infusion was increased at 40 ng/ml per 15 min to reach a maximum of 320 ng/ml. The following measurements were made: arterial plasma S(+)-ketamine and S(+)-norketamine concentrations, heat pain intensity, electrical pain tolerance, drug high, and cardiac output. The data were modeled by using sigmoid Emax models of S(+)-ketamine concentration versus effect and S(+)-ketamine + S(+)-norketamine concentrations versus effect. Results:Sex differences observed were restricted to pharmacokinetic model parameters, with a 20% greater elimination clearance of S(+)-ketamine and S(+)-norketamine in women resulting in higher drug plasma concentrations in men. S(+)-ketamine produced profound drug high and analgesia with six times greater potency in the heat pain than the electrical pain test. After ketamine-infusion, analgesia rapidly dissipated; in the heat pain test but not the electrical pain test, analgesia was followed by a period of hyperalgesia. Over the dose range tested, ketamine produced a 40–50% increase in cardiac output. A significant consistent contribution of S(+)-norketamine to overall effect was detected for none of the outcome parameters. Conclusions:S(+)-ketamine displays clinically relevant sex differences in its pharmacokinetics. It is a potent analgesic at already low plasma concentrations, but it is associated with intense side effects.

[1]  H. Gunduz-Bruce,et al.  The acute effects of NMDA antagonism: From the rodent to the human brain , 2009, Brain Research Reviews.

[2]  A. Tort,et al.  The NMDA antagonist MK-801 induces hyperalgesia and increases CSF excitatory amino acids in rats: Reversal by guanosine , 2009, Pharmacology Biochemistry and Behavior.

[3]  P. Crooks,et al.  Effects of norketamine enantiomers in rodent models of persistent pain , 2008, Pharmacology Biochemistry and Behavior.

[4]  A. Weinbroum,et al.  Ketamine spares morphine consumption after transthoracic lung and heart surgery without adverse hemodynamic effects. , 2008, Pharmacological research.

[5]  H. Dupont,et al.  Postoperative Ketamine Administration Decreases Morphine Consumption in Major Abdominal Surgery: A Prospective, Randomized, Double-Blind, Controlled Study , 2008, Anesthesia and analgesia.

[6]  C. Maier,et al.  [Sympathomimetic effects of low-dose S(+)-ketamine. Effect of propofol dosage]. , 2008, Der Anaesthesist.

[7]  M. Picker,et al.  Sex differences in NMDA antagonist enhancement of morphine antihyperalgesia in a capsaicin model of persistent pain: Comparisons to two models of acute pain , 2008, Pharmacology Biochemistry and Behavior.

[8]  J Lötsch,et al.  Differential Opioid Action on Sensory and Affective Cerebral Pain Processing , 2008, Clinical pharmacology and therapeutics.

[9]  M. Zenz,et al.  Sympathomimetische Effekte auch bei niedriger Dosierung von Esketamin , 2008, Der Anaesthesist.

[10]  N. Holford,et al.  Investigating the pharmacodynamics of ketamine in children , 2007, Paediatric anaesthesia.

[11]  N. Holford,et al.  Modeling the norketamine metabolite in children and the implications for analgesia , 2007, Paediatric anaesthesia.

[12]  W. Buhre,et al.  Validation of a new arterial pulse contour-based cardiac output device , 2007, Critical care medicine.

[13]  M O Karlsson,et al.  Diagnosing Model Diagnostics , 2007, Clinical pharmacology and therapeutics.

[14]  B. Chizh Low dose ketamine: a therapeutic and research tool to explore N-methyl-D-aspartate (NMDA) receptor-mediated plasticity in pain pathways , 2007, Journal of psychopharmacology.

[15]  Till Sprenger,et al.  Imaging Pain Modulation by Subanesthetic S-(+)-Ketamine , 2006, Anesthesia and analgesia.

[16]  J. Krystal,et al.  Greater vulnerability to the amnestic effects of ketamine in males , 2006, Psychopharmacology.

[17]  M. White,et al.  Pharmacokinetics of S(+) ketamine derived from target controlled infusion. , 2006, British journal of anaesthesia.

[18]  Erik Olofsen,et al.  Alfentanil and Placebo Analgesia: No Sex Differences Detected in Models of Experimental Pain , 2005, Anesthesiology.

[19]  M. Bushnell,et al.  The effects of racemic ketamine on painful stimulation of skin and viscera in human subjects , 2005, Pain.

[20]  Paul F. White,et al.  Pharmacodynamic modeling of the EEG effects of ketamine and its enantiomers in man , 1987, Journal of Pharmacokinetics and Biopharmaceutics.

[21]  A. Dahan,et al.  Pharmacokinetic-Pharmacodynamic Modeling of Morphine-6-glucuronide-induced Analgesia in Healthy Volunteers: Absence of Sex Differences , 2004, Anesthesiology.

[22]  Hiroshi Baba,et al.  The Role of N-Methyl-d-Aspartate (NMDA) Receptors in Pain: A Review , 2003, Anesthesia and analgesia.

[23]  J. Schüttler,et al.  Differential Modulation of Remifentanil-induced Analgesia and Postinfusion Hyperalgesia by S-Ketamine and Clonidine in Humans , 2003, Anesthesiology.

[24]  T. Iga,et al.  Plasma concentration profiles of ketamine and norketamine after administration of various ketamine preparations to healthy Japanese volunteers , 2003, Biopharmaceutics & drug disposition.

[25]  Y. Hijazi,et al.  Contribution of CYP3A4, CYP2B6, and CYP2C9 isoforms to N-demethylation of ketamine in human liver microsomes. , 2002, Drug metabolism and disposition: the biological fate of chemicals.

[26]  E. Pfenninger,et al.  Cognitive Impairment after Small-dose Ketamine Isomers in Comparison to Equianalgesic Racemic Ketamine in Human Volunteers , 2002, Anesthesiology.

[27]  H. Scheinin,et al.  Pharmacokinetics and non-analgesic effects of S- and R-ketamines in healthy volunteers with normal and reduced metabolic capacity , 2002, European Journal of Clinical Pharmacology.

[28]  T. Yaksh,et al.  Concentration‐Effect Relationships for Intravenous Alfentanil and Ketamine Infusions in Human Volunteers: Effects on Acute Thresholds and Capsaicin‐Evoked Hyperpathia , 2002, Journal of clinical pharmacology.

[29]  A. Dahan,et al.  The Involvement of the &mgr;-Opioid Receptor in Ketamine-Induced Respiratory Depression and Antinociception , 2001, Anesthesia and analgesia.

[30]  J. Schüttler,et al.  Stereoselective pharmacokinetics of ketamine: R(−)‐Ketamine inhibits the elimination of S(+)‐ketamine , 2001, Clinical pharmacology and therapeutics.

[31]  Jürgen Schüttler,et al.  A New Model of Electrically Evoked Pain and Hyperalgesia in Human Skin: The Effects of Intravenous Alfentanil, S (+)-ketamine, and Lidocaine , 2001, Anesthesiology.

[32]  T. Yaksh,et al.  Concentration–effect relationship of intravenous alfentanil and ketamine on peripheral neurosensory thresholds, allodynia and hyperalgesia of neuropathic pain , 2001, Pain.

[33]  A. Dahan,et al.  Sex Differences in Morphine Analgesia: An Experimental Study in Healthy Volunteers , 2000, Anesthesiology.

[34]  R. Likar,et al.  The effects of intradermal fentanyl and ketamine on capsaicin-induced secondary hyperalgesia and flare reaction. , 1999, Anesthesia and analgesia.

[35]  A. Sandler,et al.  Use and efficacy of low-dose ketamine in the management of acute postoperative pain: a review of current techniques and outcomes , 1999, Pain.

[36]  A. Mitchell Generalized hyperalgesia and allodynia following abrupt cessation of subcutaneous ketamine infusion , 1999, Palliative medicine.

[37]  A. Gorman,et al.  Oral ketamine is antinociceptive in the rat formalin test: role of the metabolite, norketamine , 1999, Pain.

[38]  F. M. Borgbjerg,et al.  Norketamine, the main metabolite of ketamine, is a non-competitive NMDA receptor antagonist in the rat cortex and spinal cord. , 1997, European journal of pharmacology.