Fortschr Neurol Psychiatr 2007; 75(11): 641-652
DOI: 10.1055/s-2007-959181
Originalarbeit
© Georg Thieme Verlag Stuttgart · New York

Neue Wege in der Depressionsbehandlung

New Options in the Treatment of DepressionH.  Frieling1 , T.  Hillemacher1 , J.  H.  Demling1 , J.  Kornhuber1 , S.  Bleich1
  • 1Psychiatrische und Psychotherapeutische Klinik, Universitätsklinikum Erlangen (Prof. J. Kornhuber)
Further Information

Publication History

Publication Date:
16 March 2007 (online)

Zusammenfassung

Die pharmakologische Behandlung der Depression ist trotz der enormen Fortschritte der letzten 40 Jahre noch immer nicht zufrieden stellend. Alle derzeit verwendeten Substanzen wirken über die Beeinflussung der Monoamine im zentralen Nervensystem. In der vorliegenden Übersicht sollen neben Fortentwicklungen der monoaminergen Antidepressiva neue antidepressive Wirkmechanismen vorgestellt und diskutiert werden. Neuere Entwicklungen im Bereich der monoaminergen Substanzen umfassen Metabolite gängiger Antidepressiva, direkte Serotoninagonisten oder Dreifachwiederaufnahmehemmer, die den Transport von Serotonin, Noradrenalin und Dopamin hemmen. Bei den nicht monoaminergen Strategien stehen Substanzen im Vordergrund, die auf Melatonin wirken oder Neuropeptide beeinflussen. Eine hoffnungsvolle Klasse von möglichen Antidepressiva stellen die glutamat-modulierenden Substanzen wie Ketamin und Riluzol dar. Daneben sind auch Fortschritte im Bereich der Modulierung der HPA-Achse zu verzeichnen.

Abstract

Despite the advances in the psychopharmacology, the treatment of depressive disorder is still not satisfactory. All current pharmacological substances are affecting the monoamines in the central nervous system. The present review discusses advances in the field of monoaminergic antidepressants as well as new treatment alternatives. The new monoaminergic substances include metabolites of known antidepressants, direct serotonin-agonists, and triple-reuptake inhibitors, blocking the transport of serotonin, norepinephrine, and dopamine.
Non-monoaminergic strategies include substances affecting melatonin or neuropeptides. Glutamate modulating agents such as ketamine or riluzole are another promising approach in the treatment of depression. Some advances have also been achieved in the field of HPA-axis modulation.

Literatur

  • 1 Hasin D S, Goodwin R D, Stinson F S, Grant B F. Epidemiology of major depressive disorder: results from the National Epidemiologic Survey on Alcoholism and Related Conditions.  Arch Gen Psychiatry. 2005;  62 1097-1106
  • 2 Paykel E S, Brugha T, Fryers T. Size and burden of depressive disorders in Europe.  Eur Neuropsychopharmacol. 2005;  15 411-423
  • 3 Dohmen C, Garlip G, Sitzer M, Siebler M, Malevani J, Kessler K R, Huff W. Post-Stroke-Depression. Algorithmus fur ein standardisiertes diagnostisches Vorgehen in der klinischen Routine.  Fortschr Neurol Psychiatr. 2006;  74 257-262
  • 4 Berton O, Nestler E J. New approaches to antidepressant drug discovery: beyond monoamines.  Nat Rev Neurosci. 2006;  7 137-151
  • 5 Svenningsson P, Chergui K, Rachleff I, Flajolet M, Zhang X, Yacoubi M E, Vaugeois J M, Nomikos G G, Greengard P. Alterations in 5-HT1B Receptor Function by p11 in Depression-Like States.  Science. 2006;  311 77-80
  • 6 Stimmel G L, Dopheide J A, Stahl S M. Mirtazapine: an antidepressant with noradrenergic and specific serotonergic effects.  Pharmacotherapy. 1997;  17 10-21
  • 7 Artigas F, Adell A, Celada P. Pindolol augmentation of antidepressant response.  Curr Drug Targets. 2006;  7 139-147
  • 8 Gardier A M, Malagie I, Trillat A C, Jacquot C, Artigas F. Role of 5-HT1A autoreceptors in the mechanism of action of serotoninergic antidepressant drugs: recent findings from in vivo microdialysis studies.  Fundam Clin Pharmacol. 1996;  10 16-27
  • 9 Kreiss D S, Lucki I. Chronic administration of the 5-HT1A receptor agonist 8-OH-DPAT differentially desensitizes 5-HT1A autoreceptors of the dorsal and median raphe nuclei.  Synapse. 1997;  25 107-116
  • 10 Bosker F J, Westerink B H, Cremers T I, Gerrits M, Hart M G van der, Kuipers S D, Pompe D  van der, Horst G J ter, Boer J A den, Korf J. Future antidepressants: what is in the pipeline and what is missing?.  CNS Drugs. 2004;  18 705-732
  • 11 Trivedi M H, Fava M, Wisniewski S R, Thase M E, Quitkin F, Warden D, Ritz L, Nierenberg A A, Lebowitz B D, Biggs M M, Luther J F, Shores-Wilson K, Rush A J. Medication augmentation after the failure of SSRIs for depression.  N Engl J Med. 2006;  354 1243-1252
  • 12 Keller M B, Ruwe F J, Janssens C J, Sitsen J M, Jokinen R, Janczewski J. Relapse prevention with gepirone ER in outpatients with major depression.  J Clin Psychopharmacol. 2005;  25 79-84
  • 13 Amsterdam J D, Brunswick D J, Gibertini M. Sustained efficacy of gepirone-IR in major depressive disorder: a double-blind placebo substitution trial.  J Psychiatr Res. 2004;  38 259-265
  • 14 Feiger A D, Heiser J F, Shrivastava R K, Weiss K J, Smith W T, Sitsen J M, Gibertini M. Gepirone extended-release: new evidence for efficacy in the treatment of major depressive disorder.  J Clin Psychiatry. 2003;  64 243-249
  • 15 Heinrich T, Bottcher H, Gericke R, Bartoszyk G D, Anzali S, Seyfried C A, Greiner H E, Amsterdam C Van. Synthesis and structure - activity relationship in a class of indolebutylpiperazines as dual 5-HT(1A) receptor agonists and serotonin reuptake inhibitors.  J Med Chem. 2004;  47 4684-4692
  • 16 Roberts C, Hagan J J, Bartoszyk G D, Kew J N. Effect of vilazodone on 5-HT efflux and re-uptake in the guinea-pig dorsal raphe nucleus.  Eur J Pharmacol. 2005;  517 59-63
  • 17 Storosum J G, Elferink A J, Zwieten B J van, Block B W van den, Huyser J. Natural course and placebo response in short-term, placebo-controlled studies in major depression: a meta-analysis of published and non-published studies.  Pharmacopsychiatry. 2004;  37 32-36
  • 18 Schechter L E, Ring R H, Beyer C E, Hughes Z A, Khawaja X, Malberg J E, Rosenzweig-Lipson S. Innovative approaches for the development of antidepressant drugs: current and future strategies.  NeuroRx. 2005;  2 590-611
  • 19 Simiand J, Keane P E, Guitard J, Langlois X, Gonalons N, Martin P, Bianchetti A, Le Fur G, Soubrie P. Antidepressant profile in rodents of SR 58611A, a new selective agonist for atypical beta-adrenoceptors.  Eur J Pharmacol. 1992;  219 193-201
  • 20 ADIS International R&D profile . SR 58611A: SR 58 611.  Drugs R D. 2003;  4 380-382
  • 21 Halaris A E, Belendiuk K T, Freedman D X. Antidepressant drugs affect dopamine uptake.  Biochem Pharmacol. 1975;  24 1896-1897
  • 22 Randrup A, Braestrup C. Uptake inhibition of biogenic amines by newer antidepressant drugs: relevance to the dopamine hypothesis of depression.  Psychopharmacology (Berl). 1977;  53 309-314
  • 23 DeBattista C, Lembke A. Update on augmentation of antidepressant response in resistant depression.  Curr Psychiatry Rep. 2005;  7 435-440
  • 24 Ascher J A, Cole J O, Colin J N, Feighner J P, Ferris R M, Fibiger H C, Golden R N, Martin P, Potter W Z, Richelson E. Bupropion: a review of its mechanism of antidepressant activity.  J Clin Psychiatry. 1995;  56 395-401
  • 25 Gross G, Xin X, Gastpar M. Trimipramine: pharmacological reevaluation and comparison with clozapine.  Neuropharmacology. 1991;  30 1159-1166
  • 26 Piercey M F. Pharmacology of pramipexole, a dopamine D3-preferring agonist useful in treating Parkinson's disease.  Clin Neuropharmacol. 1998;  21 141-151
  • 27 Rektorova I, Rektor I, Bares M, Dostal V, Ehler E, Fanfrdlova Z, Fiedler J, Klajblova H, Kulist'ak P, Ressner P, Svatova J, Urbanek K, Veliskova J. Pramipexole and pergolide in the treatment of depression in Parkinson's disease: a national multicentre prospective randomized study.  Eur J Neurol. 2003;  10 399-406
  • 28 Corrigan M H, Denahan A Q, Wright C E, Ragual R J, Evans D L. Comparison of pramipexole, fluoxetine, and placebo in patients with major depression.  Depress Anxiety. 2000;  11 58-65
  • 29 Goldberg J F, Burdick K E, Endick C J. Preliminary randomized, double-blind, placebo-controlled trial of pramipexole added to mood stabilizers for treatment-resistant bipolar depression.  Am J Psychiatry. 2004;  161 564-566
  • 30 Lattanzi L, Dell'Osso L, Cassano P, Pini S, Rucci P, Houck P R, Gemignani A, Battistini G, Bassi A, Abelli M, Cassano G B. Pramipexole in treatment-resistant depression: a 16-week naturalistic study.  Bipolar Disord. 2002;  4 307-314
  • 31 Goldberg J F, Frye M A, Dunn R T. Pramipexole in refractory bipolar depression.  Am J Psychiatry. 1999;  156 798
  • 32 Cassano P, Lattanzi L, Soldani F, Navari S, Battistini G, Gemignani A, Cassano G B. Pramipexole in treatment-resistant depression: an extended follow-up.  Depress Anxiety. 2004;  20 131-138
  • 33 Rocca P, Fonzo V, Ravizza L, Rocca G, Scotta M, Zanalda E, Bogetto F. A comparison of paroxetine and amisulpride in the treatment of dysthymic disorder.  J Affect Disord. 2002;  70 313-317
  • 34 Cassano G B, Jori M C. Efficacy and safety of amisulpride 50 mg versus paroxetine 20 mg in major depression: a randomized, double-blind, parallel group study.  Int Clin Psychopharmacol. 2002;  17 27-32
  • 35 Amore M, Jori M C. Faster response on amisulpride 50 mg versus sertraline 50 - 100 mg in patients with dysthymia or double depression: a randomized, double-blind, parallel group study.  Int Clin Psychopharmacol. 2001;  16 317-324
  • 36 Schmauss M, Messer T. Dysthymie.  Fortschr Neurol Psychiatr. 2005;  73 415-424
  • 37 Mucci A, Nolfe G, Maj M. Levosulpiride: a review of its clinical use in psychiatry.  Pharmacol Res. 1995;  31 95-101
  • 38 Amsterdam J D, Brunswick D J, Hundert M. A single-site, double-blind, placebo-controlled, dose-ranging study of YKP10A - a putative, new antidepressant.  Prog Neuropsychopharmacol Biol Psychiatry. 2002;  26 1333-1338
  • 39 Skolnick P, Popik P, Janowsky A, Beer B, Lippa A S. Antidepressant-like actions of DOV 21,947: a „triple” reuptake inhibitor.  Eur J Pharmacol. 2003;  461 99-104
  • 40 FDA . First depression patch approved.  FDA Consum. 2006;  40 4
  • 41 Wessel K, Szelenyi I. Selegiline - an overview of its role in the treatment of Parkinson's disease.  Clin Investig. 1992;  70 459-462
  • 42 Benedictis E. 20 mg transdermal selegiline daily may be effective and well tolerated in adults with major depression.  Evid Based Ment Health. 2003;  6 44
  • 43 Amsterdam J D. A double-blind, placebo-controlled trial of the safety and efficacy of selegiline transdermal system without dietary restrictions in patients with major depressive disorder.  J Clin Psychiatry. 2003;  64 208-214
  • 44 Bodkin J A, Amsterdam J D. Transdermal selegiline in major depression: a double-blind, placebo-controlled, parallel-group study in outpatients.  Am J Psychiatry. 2002;  159 1869-1875
  • 45 Frieling H, Bleich S. Tranylcypromine: New perspectives on an “old” drug.  Eur Arch Psychiatry Clin Neurosci. 2006;  256 268-273
  • 46 Yildiz-Yesiloglu A, Ankerst D P. Review of (1)H magnetic resonance spectroscopy findings in major depressive disorder: A meta-analysis.  Psychiatry Res. 2006;  147 1-25
  • 47 Bjelland I, Tell G S, Vollset S E, Refsum H, Ueland P M. Folate, vitamin B12, homocysteine, and the MTHFR 677C->T polymorphism in anxiety and depression: the Hordaland Homocysteine Study.  Arch Gen Psychiatry. 2003;  60 618-626
  • 48 Goodwin G M, Bowden C L, Calabrese J R, Grunze H, Kasper S, White R, Greene P, Leadbetter R. A pooled analysis of 2 placebo-controlled 18-month trials of lamotrigine and lithium maintenance in bipolar I disorder.  J Clin Psychiatry. 2004;  65 432-441
  • 49 Nowak G, Trullas R, Layer R T, Skolnick P, Paul I A. Adaptive changes in the N-methyl-D-aspartate receptor complex after chronic treatment with imipramine and 1-aminocyclopropanecarboxylic acid.  J Pharmacol Exp Ther. 1993;  265 1380-1386
  • 50 Zarate Jr C A, Singh J B, Carlson P J, Brutsche N E, Ameli R, Luckenbaugh D A, Charney D S, Manji H K. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression.  Arch Gen Psychiatry. 2006;  63 856-864
  • 51 Kornhuber J, Bormann J, Retz W, Hubers M, Riederer P. Memantine displaces [3H]MK-801 at therapeutic concentrations in postmortem human frontal cortex.  Eur J Pharmacol. 1989;  166 589-590
  • 52 Zarate Jr C A, Singh J B, Quiroz J A, DeJesus G, Denicoff K K, Luckenbaugh D A, Manji H K, Charney D S. A double-blind, placebo-controlled study of memantine in the treatment of major depression.  Am J Psychiatry. 2006;  163 153-155
  • 53 Kos T, Legutko B, Danysz W, Samoriski G, Popik P. Enhancement of Antidepressant-Like Effects but Not Brain-Derived Neurotrophic Factor mRNA Expression by the Novel N-Methyl-D-aspartate Receptor Antagonist Neramexane in Mice.  J Pharmacol Exp Ther. 2006;  318 1128-1136
  • 54 Mathew S J, Keegan K, Smith L. Glutamate modulators as novel interventions for mood disorders.  Rev Bras Psiquiatr. 2005;  27 243-248
  • 55 Borges K, Dingledine R. AMPA receptors: molecular and functional diversity.  Prog Brain Res. 1998;  116 153-170
  • 56 Goff D C, Leahy L, Berman I, Posever T, Herz L, Leon A C, Johnson S A, Lynch G. A placebo-controlled pilot study of the ampakine CX516 added to clozapine in schizophrenia.  J Clin Psychopharmacol. 2001;  21 484-487
  • 57 Li X, Tizzano J P, Griffey K, Clay M, Lindstrom T, Skolnick P. Antidepressant-like actions of an AMPA receptor potentiator (LY392098).  Neuropharmacology. 2001;  40 1028-1033
  • 58 Knapp R J, Goldenberg R, Shuck C, Cecil A, Watkins J, Miller C, Crites G, Malatynska E. Antidepressant activity of memory-enhancing drugs in the reduction of submissive behavior model.  Eur J Pharmacol. 2002;  440 27-35
  • 59 Lauterborn J C, Truong G S, Baudry M, Bi X, Lynch G, Gall C M. Chronic elevation of brain-derived neurotrophic factor by ampakines.  J Pharmacol Exp Ther. 2003;  307 297-305
  • 60 Lauterborn J C, Lynch G, Vanderklish P, Arai A, Gall C M. Positive modulation of AMPA receptors increases neurotrophin expression by hippocampal and cortical neurons.  J Neurosci. 2000;  20 8-21
  • 61 Thome J, Bleich S. Molekularbiologische Grundlagen antidepressiver Therapie. In: Demling J (Hrsg). Therapieresistente Depressionen. Bremen; London; Boston: UNI-MED Verlag AG 2004: 80-87
  • 62 Danysz W. LY-544 344. Eli Lilly.  IDrugs. 2005;  8 755-762
  • 63 Wang S J, Wang K Y, Wang W C. Mechanisms underlying the riluzole inhibition of glutamate release from rat cerebral cortex nerve terminals (synaptosomes).  Neuroscience. 2004;  125 191-201
  • 64 Du J, Suzuki K, Wei Y, Wang Y, Blumenthal R, Chen Z, Falke C, Zarate Jr C A, Manji H K. The Anticonvulsants Lamotrigine, Riluzole, and Valproate Differentially Regulate AMPA Receptor Membrane Localization: Relationship to Clinical Effects in Mood Disorders.  Neuropsychopharmacology. 2006;  doi: 10.1038/sj.npp.1301178
  • 65 Zarate Jr C A, Payne J L, Quiroz J, Sporn J, Denicoff K K, Luckenbaugh D, Charney D S, Manji H K. An open-label trial of riluzole in patients with treatment-resistant major depression.  Am J Psychiatry. 2004;  161 171-174
  • 66 Zarate Jr C A, Quiroz J A, Singh J B, Denicoff K D, DeJesus G, Luckenbaugh D A, Charney D S, Manji H K. An open-label trial of the glutamate-modulating agent riluzole in combination with lithium for the treatment of bipolar depression.  Biol Psychiatry. 2005;  57 430-432
  • 67 Sanacora G, Kendell S F, Fenton L, Coric V, Krystal J H. Riluzole augmentation for treatment-resistant depression.  Am J Psychiatry. 2004;  161 2132
  • 68 Nowak G, Partyka A, Palucha A, Szewczyk B, Wieronska J M, Dybala M, Metz M, Librowski T, Froestl W, Papp M, Pilc A. Antidepressant-like activity of CGP 36 742 and CGP 51 176, selective GABA(B) receptor antagonists, in rodents.  Br J Pharmacol. 2006;  149 581-590
  • 69 Ong J, Kerr D. Clinical potential of receptor modulators.  CNS Drug Rev. 2005;  11 317-334
  • 70 Pilc A, Nowak G. GABAergic hypotheses of anxiety and depression: focus on GABA-B receptors.  Drugs Today (Barc ). 2005;  41 755-766
  • 71 Vieta E, Manuel G J, Martinez-Aran A, Comes M, Verger K, Masramon X, Sanchez-Moreno J, Colom F. A double-blind, randomized, placebo-controlled, prophylaxis study of adjunctive gabapentin for bipolar disorder.  J Clin Psychiatry. 2006;  67 473-477
  • 72 Carpenter L L, Schecter J M, Tyrka A R, Mello A F, Mello M F, Haggarty R, Price L. Open-label tiagabine monotherapy for major depressive disorder with anxiety.  J Clin Psychiatry. 2006;  67 66-71
  • 73 Ebert D, Kaschka W. Humorale Aspekte des Schlafentzuges.  Fortschr Neurol Psychiatr. 1995;  63 441-450
  • 74 Melatonin. Monograph.  Altern Med Rev. 2005;  10 326-336
  • 75 Pacchierotti C, Iapichino S, Bossini L, Pieraccini F, Castrogiovanni P. Melatonin in Psychiatric Disorders: A Review on the Melatonin Involvement in Psychiatry.  Frontiers in Neuroendocrinology. 2001;  22 18-32
  • 76 Beck-Friis J, Kjellman B F, Aperia B, Unden F, Rosen D von, Ljunggren J G, Wetterberg L. Serum melatonin in relation to clinical variables in patients with major depressive disorder and a hypothesis of a low melatonin syndrome.  Acta Psychiatr Scand. 1985;  71 319-330
  • 77 Brown R, Kocsis J H, Caroff S, Amsterdam J, Winokur A, Stokes P E, Frazer A. Differences in nocturnal melatonin secretion between melancholic depressed patients and control subjects.  Am J Psychiatry. 1985;  142 811-816
  • 78 Boer J A den, Bosker F J, Meesters Y. Clinical efficacy of agomelatine in depression: the evidence.  Int Clin Psychopharmacol. 2006;  21 Suppl 1 S21-S24
  • 79 Kennedy S H, Emsley R. Placebo-controlled trial of agomelatine in the treatment of major depressive disorder.  Eur Neuropsychopharmacol. 2006;  16 93-100
  • 80 Tuma J, Strubbe J H, Mocaer E, Koolhaas J M. Anxiolytic-like action of the antidepressant agomelatine (S 20 098) after a social defeat requires the integrity of the SCN.  Eur Neuropsychopharmacol. 2005;  15 545-555
  • 81 Barden N, Shink E, Labbe M, Vacher R, Rochford J, Mocaer E. Antidepressant action of agomelatine (S 20 098) in a transgenic mouse model.  Prog Neuropsychopharmacol Biol Psychiatry. 2005;  29 908-916
  • 82 Loiseau F, Le Bihan C, Hamon M, Thiebot M H. Antidepressant-like effects of agomelatine, melatonin and the NK1 receptor antagonist GR205171 in impulsive-related behaviour in rats.  Psychopharmacology (Berl). 2005;  182 24-32
  • 83 Bourin M, Mocaer E, Porsolt R. Antidepressant-like activity of S 20 098 (agomelatine) in the forced swimming test in rodents: involvement of melatonin and serotonin receptors.  J Psychiatry Neurosci. 2004;  29 126-133
  • 84 Papp M, Gruca P, Boyer P A, Mocaer E. Effect of agomelatine in the chronic mild stress model of depression in the rat.  Neuropsychopharmacology. 2003;  28 694-703
  • 85 Loo H, Dalery J, Macher J P, Payen A. Etude pilote comparant en aveugle l'effet therapeutique de deux doses d'agomelatine - agoniste des recepteurs de la melatonine et antagoniste des recepteurs 5HT2c - chez 30 patients.  Encephale. 2002;  28 356-362
  • 86 Loo H, Hale A, D'haenen H. Determination of the dose of agomelatine, a melatoninergic agonist and selective 5-HT(2C) antagonist, in the treatment of major depressive disorder: a placebo-controlled dose range study.  Int Clin Psychopharmacol. 2002;  17 239-247
  • 87 Montgomery S A, Kennedy S H, Burrows G D, Lejoyeux M, Hindmarch I. Absence of discontinuation symptoms with agomelatine and occurrence of discontinuation symptoms with paroxetine: a randomized, double-blind, placebo-controlled discontinuation study.  Int Clin Psychopharmacol. 2004;  19 271-280
  • 88 Papp M, Litwa E, Gruca P, Mocaer E. Anxiolytic-like activity of agomelatine and melatonin in three animal models of anxiety.  Behav Pharmacol. 2006;  17 9-18
  • 89 McGechan A, Wellington K. Ramelteon.  CNS Drugs. 2005;  19 1057-1065
  • 90 Prestele S, Aldenhoff J, Reiff J. Die HPA-Achse als mogliches Bindeglied zwischen Depression, Diabetes mellitus und kognitiven Störungen.  Fortschr Neurol Psychiatr. 2003;  71 24-36
  • 91 Schmider J, Lammers C H, Gotthardt U, Dettling M, Holsboer F, Heuser I J. Combined dexamethasone/corticotropin-releasing hormone test in acute and remitted manic patients, in acute depression, and in normal controls: I.  Biol Psychiatry. 1995;  38 797-802
  • 92 Sharma R, Hedeker D, Pandey G, Janicak P, Davis J. A longitudinal study of plasma cortisol and depressive symptomatology by random regression analysis.  Biol Psychiatry. 1992;  31 304-314
  • 93 Szyf M, Weaver I CG, Champagne F A, Diorio J, Meaney M J. Maternal programming of steroid receptor expression and phenotype through DNA methylation in the rat.  Frontiers in Neuroendocrinology. 2005;  26 139-162
  • 94 Kunzel H E, Ising M, Zobel A W, Nickel T, Ackl N, Sonntag A, Holsboer F, Uhr M. Treatment with a CRH-1-receptor antagonist (R121919) does not affect weight or plasma leptin concentration in patients with major depression.  Journal of Psychiatric Research. 2005;  39 173-177
  • 95 Zobel A W, Nickel T, Kunzel H E, Ackl N, Sonntag A, Ising M, Holsboer F. Effects of the high-affinity corticotropin-releasing hormone receptor 1 antagonist R121919 in major depression: the first 20 patients treated.  Journal of Psychiatric Research. 2000;  34 171-181
  • 96 DeBattista C, Belanoff J, Glass S, Khan A, Horne R L, Blasey C, Carpenter L L, Alva G. Mifepristone versus Placebo in the Treatment of Psychosis in Patients with Psychotic Major Depression.  Biol Psychiatry. 2006;  60 1343-1349
  • 97 Mayer J L, Klumpers L, Maslam S, de Kloet E R, Joels M, Lucassen P J. Brief treatment with the glucocorticoid receptor antagonist mifepristone normalises the corticosterone-induced reduction of adult hippocampal neurogenesis.  J Neuroendocrinol. 2006;  18 629-631
  • 98 Jahn H, Schick M, Kiefer F, Kellner M, Yassouridis A, Wiedemann K. Metyrapone as additive treatment in major depression: a double-blind and placebo-controlled trial.  Arch Gen Psychiatry. 2004;  61 1235-1244
  • 99 Serradeil-Le Gal C, Derick S, Brossard G, Manning M, Simiand J, Gaillard R, Griebel G, Guillon G. Functional and pharmacological characterization of the first specific agonist and antagonist for the V1b receptor in mammals.  Stress. 2003;  6 199-206
  • 100 Cirillo R, Gillio T E, Schwarz M K, Quattropani A, Scheer A, Missotten M, Dorbais J, Nichols A, Borrelli F, Giachetti C, Golzio L, Marinelli P, Thomas R, Chevillard C, Laurent F, Portet K, Barberis C, Chollet A. Pharmacology of (2S,4Z)-N-[(2S)-2-hydroxy-2-phenylethyl]-4-(methoxyimino) -1-[(2’-methyl[1,1’-biphenyl]-4-yl)carbonyl]-2-pyrrolidinecarboxamide, a new potent and selective nonpeptide antagonist of the oxytocin receptor.  J Pharmacol Exp Ther. 2003;  306 253-261
  • 101 Griebel G, Stemmelin J, Gal C S, Soubrie P. Non-peptide vasopressin V1b receptor antagonists as potential drugs for the treatment of stress-related disorders.  Curr Pharm Des. 2005;  11 1549-1559
  • 102 Overstreet D H, Hlavka J, Feighner J P, Nicolau G, Freed J S. Antidepressant-like effects of a novel pentapeptide, nemifitide, in an animal model of depression.  Psychopharmacology (Berl). 2004;  175 303-309
  • 103 Maggi C A. The mammalian tachykinin receptors.  Gen Pharmacol. 1995;  26 911-944
  • 104 Herpfer I, Lieb K. Substance P receptor antagonists in psychiatry: rationale for development and therapeutic potential.  CNS Drugs. 2005;  19 275-293
  • 105 Rupniak N M, Kramer M S. Discovery of the antidepressant and anti-emetic efficacy of substance P receptor (NK1) antagonists.  Trends Pharmacol Sci. 1999;  20 485-490
  • 106 Ranga K, Krishnan R. Clinical experience with substance P receptor (NK1) antagonists in depression.  J Clin Psychiatry. 2002;  63 Suppl 11 25-29
  • 107 Keller M, Montgomery S, Ball W, Morrison M, Snavely D, Liu G, Hargreaves R, Hietala J, Lines C, Beebe K, Reines S. Lack of Efficacy of the Substance P (Neurokinin1 Receptor) Antagonist Aprepitant in the Treatment of Major Depressive Disorder.  Biological Psychiatry. 2006;  59 216-223
  • 108 Rupniak N M. New insights into the antidepressant actions of substance P (NK1 receptor) antagonists.  Can J Physiol Pharmacol. 2002;  80 489-494
  • 109 Steinberg R, Alonso R, Griebel G, Bert L, Jung M, Oury-Donat F, Poncelet M, Gueudet C, Desvignes C, Le Fur G, Soubrie P. Selective blockade of neurokinin-2 receptors produces antidepressant-like effects associated with reduced corticotropin-releasing factor function.  J Pharmacol Exp Ther. 2001;  299 449-458
  • 110 Kalra S P, Kalra P S. NPY - an endearing journey in search of a neurochemical on/off switch for appetite, sex and reproduction.  Peptides. 2004;  25 465-471
  • 111 Jimenez-Vasquez P A, Diaz-Cabiale Z, Caberlotto L, Bellido I, Overstreet D, Fuxe K, Mathe A A. Electroconvulsive stimuli selectively affect behavior and neuropeptide Y (NPY) and NPY Y(1) receptor gene expressions in hippocampus and hypothalamus of Flinders Sensitive Line rat model of depression.  Eur Neuropsychopharmacol. 2007;  17 298-308
  • 112 Karlsson R M, Holmes A, Heilig M, Crawley J N. Anxiolytic-like actions of centrally-administered neuropeptide Y, but not galanin, in C57BL/6J mice.  Pharmacol Biochem Behav. 2005;  80 427-436
  • 113 Husum H, Mikkelsen J D, Hogg S, Mathe A A, Mork A. Involvement of hippocampal neuropeptide Y in mediating the chronic actions of lithium, electroconvulsive stimulation and citalopram.  Neuropharmacology. 2000;  39 1463-1473
  • 114 Tsankova N M, Berton O, Renthal W, Kumar A, Neve R L, Nestler E J. Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action.  Nat Neurosci. 2006;  9 519-525
  • 115 Tsankova N M, Kumar A, Nestler E J. Histone modifications at gene promoter regions in rat hippocampus after acute and chronic electroconvulsive seizures.  J Neurosci. 2004;  24 5603-5610
  • 116 Abdolmaleky H M, Smith C L, Faraone S V, Shafa R, Stone W, Glatt S J, Tsuang M. Methylomics in psychiatry: Modulation of gene-environment interactions may be through DNA methylation.  Am J Med Genet B Neuropsychiatr Genet. 2004;  127 51-59
  • 117 Di Marzo V, De Petrocellis L. Plant, synthetic, and endogenous cannabinoids in medicine.  Annual Review of Medicine. 2006;  57 553-574
  • 118 Pi-Sunyer F X, Aronne L J, Heshmati H M, Devin J, Rosenstock J. for the RIO-North America Study Group . Effect of Rimonabant, a Cannabinoid-1 Receptor Blocker, on Weight and Cardiometabolic Risk Factors in Overweight or Obese Patients: RIO-North America: A Randomized Controlled Trial.  JAMA. 2006;  295 761-775
  • 119 Kornhuber J, Medlin A, Bleich S, Jendrossek V, Henkel A W, Wiltfang J, Gulbins E. High activity of acid sphingomyelinase in major depression.  J Neural Transm. 2005;  112 1583-1590
  • 120 Kornhuber J, Herr B, Thome J, Riederer P. The antiparkinsonian drug budipine binds to NMDA and sigma receptors in postmortem human brain tissue.  J Neural Transm Suppl. 1995;  46 131-137
  • 121 Kornhuber J, Schoppmeyer K, Riederer P. Affinity of 1-aminoadamantanes for the sigma binding site in post-mortem human frontal cortex.  Neurosci Lett. 1993;  163 129-131
  • 122 Akunne H C, Zoski K T, Whetzel S Z, Cordon J J, Brandon R M, Roman F, Pugsley T A. Neuropharmacological profile of a selective sigma ligand, igmesine: a potential antidepressant.  Neuropharmacology. 2001;  41 138-149
  • 123 Volz H P, Stoll K D. Clinical trials with sigma ligands.  Pharmacopsychiatry. 2004;  37 Suppl 3 S214-S220
  • 124 Bermack J E, Debonnel G. Effects of OPC-14 523, a combined sigma and 5-HT1a ligand, on pre- and post-synaptic 5-HT1a receptors.  J Psychopharmacol. 2007;  21 85-92
  • 125 Wong M L, Licinio J. From monoamines to genomic targets: a paradigm shift for drug discovery in depression.  Nat Rev Drug Discov. 2004;  3 136-151
  • 126 Linden M. Leitlinien und die Psychologie medizinischer Entscheidungsprozesse bei der Behandlung depressiver Erkrankungen.  Fortschr Neurol Psychiatr. 2005;  73 249-258

Dr. med. Helge Frieling

Psychiatrische und Psychotherapeutische Klinik Universitätsklinikum Erlangen

Schwabachanlage 6

91054 Erlangen

Email: helge.frieling@uk-erlangen.de

    >