Download True Remission in Depression: The Ultimate Goal

Document related concepts

Political abuse of psychiatry wikipedia , lookup

Mental status examination wikipedia , lookup

Mania wikipedia , lookup

Conversion disorder wikipedia , lookup

Critical Psychiatry Network wikipedia , lookup

Dissociative identity disorder wikipedia , lookup

History of mental disorders wikipedia , lookup

Dysthymia wikipedia , lookup

Pyotr Gannushkin wikipedia , lookup

Child psychopathology wikipedia , lookup

Generalized anxiety disorder wikipedia , lookup

Emergency psychiatry wikipedia , lookup

Moral treatment wikipedia , lookup

History of psychiatric institutions wikipedia , lookup

Abnormal psychology wikipedia , lookup

Postpartum depression wikipedia , lookup

Bipolar II disorder wikipedia , lookup

History of psychiatry wikipedia , lookup

Behavioral theories of depression wikipedia , lookup

Controversy surrounding psychiatry wikipedia , lookup

Epigenetics of depression wikipedia , lookup

Major depressive disorder wikipedia , lookup

Antidepressant wikipedia , lookup

Biology of depression wikipedia , lookup

Depression in childhood and adolescence wikipedia , lookup

Transcript
I S S U E
99
Medicographia
Vol 31, No. 2, 2009
ISSN 0243-3397
T rue R emission in D epression:
T he U ltimate G oal
J. MENDLEWICZ,
BELGIUM
EDITORIAL
DEFINING REMISSION IN DEPRESSION: THE CHALLENGE 113
OF COMPLETE RECOVERY.
DÉFINIR LA RÉMISSION DANS LA DÉPRESSION :
LE DÉFI DE LA GUÉRISON COMPLÈTE
H.-J. MÖLLER,
F. H. SEEMÜLLER, AND
M. RIEDEL, GERMANY
TIME COURSE OF RESPONSE AND REMISSION DURING
D. S. BALDWIN AND
A. T. V. LOPES,
UNITED KINGDOM
THE INFLUENCE OF COMORBID ANXIETY DISORDERS
P. MONTELEONE AND
M. MAJ, ITALY
CIRCADIAN RHYTHM DISTURBANCES IN DEPRESSION:
118
ANTIDEPRESSANT TREATMENT
126
ON OUTCOME IN MAJOR DEPRESSIVE DISORDER
132
IMPLICATIONS FOR TREATMENT AND QUALITY OF
REMISSION
C. R. SOLDATOS AND
IMPROVEMENT OF THE SLEEP-WAKE CYCLE AS A
C. G. THELERITIS, GREECE TARGET FOR REMISSION IN DEPRESSION
S. H. KENNEDY AND
S. RIZVI, CANADA
A journal of
medical information
and international
communication
from Servier
Available online at
www.medicographia.com
SEXUAL FUNCTIONING AND QUALITY OF LIFE IN
146
PATIENTS IN REMISSION FROM DEPRESSION
J. PRICE AND G. M. GOOD- EMOTIONAL BLUNTING OR REDUCED REACTIVITY
FOLLOWING REMISSION OF MAJOR DEPRESSION
WIN, UNITED KINGDOM
E. S. PAYKEL,
UNITED KINGDOM
140
RESIDUAL SYMPTOMS AND RELAPSE IN DEPRESSION
152
157
Contents continued overleaf...
Medicographia
Vol 31, No. 2, 2009
I S S U E
99
...Contents continued from cover page
T rue R emission in D epression:
T he U ltimate G oal
M. JAREMA, POLAND /
M. BAUER, GERMANY /
E. SACCHETTI, ITALY /
P. A. SCHMIDT DO
PRADO-LIMA, BRAZIL /
H. ÅGREN, SWEDEN /
J. PYO HONG, KOREA /
R. MAHENDRAN, SINGAPORE / A. HATIM
SULAIMAN, MALAYSIA /
F. CAÑAS, SPAIN /
F. T. ANTUN, LEBANON
C. MUÑOZ, FRANCE
CONTROVERSIAL QUESTION
IS THE PATIENT REALLY THE SAME AFTER A MAJOR
DEPRESSIVE EPISODE?
164
VALDOXAN
BETTER QUALITY REMISSION IN DEPRESSION:
VALDOXAN, THE FIRST MELATONERGIC
175
ANTIDEPRESSANT
W. CHOUCHA AND
J.-F. ALLILAIRE, FRANCE
G. A. FAVA AND
D. VISANI, ITALY
INTERVIEW
CLINICAL MANAGEMENT OF PATIENTS FOLLOWING
182
REMISSION FROM DEPRESSION
FOCUS
INCREASING THE LEVEL OF REMISSION IN DEPRESSION
186
BY THE SEQUENTIAL USE OF PHARMACOTHERAPY
AND PSYCHOTHERAPY
J. D. GUELFI, FRANCE
UPDATE
MEASURING REMISSION IN DEPRESSION
GU QIAN, NI LIDONG,
HU WEIGUO, CHINA
A TOUCH OF FRANCE
RUIJIN HOSPITAL. FROM 19TH-CENTURY FRENCH
JESUIT HOSPITAL TO PRESENT-DAY STATE-OF-THEART CHINESE UNIVERSITY HOSPITAL
D. CAMUS, FRANCE
A TOUCH OF FRANCE
“PARIS OF THE ORIENT.” THE SHANGHAI FRENCH
CONCESSION (1849-1946)
192
198
205
E
D I T O R I A L
Defining remission in depression:
the challenge of complete recovery
by J. Mendlewicz, Belgium
D
Julien MENDLEWICZ
MD, PhD
Faculty of Medicine
Free University of Brussels
Brusssels, BELGIUM
www.medicographia.com
Address for correspondence:
Julien Mendlewicz, MD, PhD,
Full Professor of Psychiatry,
Faculty of Medicine,
Free University of Brussels,
Zonnebloemlaan, 4,
1653 Dworp-Beersel, Belgium
(e-mail:
[email protected])
Medicographia.
2009;31:113-117.
EPRESSIVE DISORDERS CONSTITUTE A MAJOR PUBLIC HEALTH
issue, and it is estimated by the World Health Organization that they
will rank in second position among all diseases in terms of prevalence by the year 2010, thus contributing heavily to the global burden of disease in humans. Advances in the treatment of depressive disorders will therefore constitute a major challenge for the medical community and for society in the years to come. The
concept of clinical remission in the treatment of major depressive disorders has gained growing
attention in the last few years. The reasons for this relatively recent interest are severalfold: depressed patients as well as patient organizations are not totally satisfied with the current effectiveness and tolerability of available antidepressant medications. Despite the obvious benefits of
antidepressants, many depressed patients still suffer from incapacitating residual symptoms, and
these patients are at a higher risk of relapse or recurrence than patients who achieve full remission after antidepressant therapy. Patients who reach full remission after treatment have a better
level of functioning and an improved prognosis compared with patients who are nonremitters.
Adequate clinical remission is therefore of great functional importance to the patient, because
it seems to be a predictor of long-term stability and a rather good indicator of better psychological functioning, the latter being of utmost importance when assessing the quality of life of our
depressed patients. For the above reasons, it is becoming of great interest to the scientific community and our patients to consider not only rates of response, but also remission rates, in order
to assess the real clinical efficiency of antidepressants in everyday practice and to evaluate new
treatments in outcome studies. This issue of Medicographia brings together experts in the field
of affective disorders to discuss from different perspectives the critical issues related to the concept of true remission in depression.
H. J. Möller et al review current knowledge on the natural course of a depressive episode, symptoms that may be predictive of response and remission, and the time course of their alleviation
during antidepressant therapy.
D. Baldwin and A. Lopes focus on the differential effects of antidepressants on endogenous depression and depression associated with anxiety disorders, in relation to the time course of depression and the quality of remission in depression.
P. Monteleone and M. Maj look at the relevance of circadian rhythm disturbances in depression
and the impact that the resynchronization of circadian rhythms has on the quality of remission.
C. Soldatos and C. Theleritis reflect on the quality of the sleep-wake cycle as a potential marker of remission in depression. They also deal with such issues as the persistence of sleep disruption after antidepressant treatment, and the promises of sleep-restorative antidepressants.
S. Kennedy and S. Rizvi emphasize the importance of the maintenance of proper sexual functioning and quality of life in patients in remission from depression, which is also a key factor for
drug compliance.
J. Price and G. Goodwin analyze the impact of antidepressant therapy on cognitive and emotional reactivity after remission and the implications this has for relapse.
Defining remission in depression: the challenge of complete recovery – Mendlewicz
MEDICOGRAPHIA, VOL 31, No. 2, 2009 113
EDITORIAL
E. Paykel draws a list of the residual symptoms whose presence signals incomplete remission
from depression and stresses the need for longer than usual continuation antidepressant treatment, which may be aided by cognitive therapy, to avoid the risk of relapse.
C. Muñoz reviews the pharmacological mode of action and clinical benefits of agomelatine, a
new melatonergic antidepressant.
W. Choucha and J. F. Allilaire discuss the various clinical management approaches to treating
remitted patients over the long term.
G. Fava and D. Visani advocate the sequential use of pharmacotherapy and psychotherapy to
address residual symptoms in patients having recovered from depression as the best model for
avoiding relapse or recurrence, and stress the implications of this method in terms of treatment
planning.
J. D. Guelfi discusses the merits and pitfalls of various methods to assess remission, relapse,
and residual symptoms in depression.
Keywords: remission;
recovery; residual
symptom; antidepressant; major depression
Definitions of remission can vary across the literature, and questions arise as to the boundary between full remission and partial remission, the presence after treatment of residual symptoms, and the return (or not) to premorbid psychosocial functioning. According to a consensus
conference of the MacArthur Foundation, clinical response is defined as a period of time during
which there is some improvement in symptoms, but not of enough magnitude as to represent
achievement of full remission, with the persistence of some residual symptoms. On depression
rating scales, this state usually corresponds to at least a 50% improvement in scores. By contrast,
full remission is obtained when clinical improvement is such that the patient becomes almost
asymptomatic. Clinical remission is usually defined by a score of 7 or less on the 17-item Hamilton Rating Scale for Depression (HAM-D17) or a score of 10 to 12 or less on the MontgomeryÅsberg Depression Rating Scale (MADRS). The persistence of remission over time while on maintenance antidepressant therapy is of obvious clinical relevance, as considered in the concept of
recovery.
Clinical recovery can only be defined in an individual after the persistence of remission for
at least 3 to 4 months. A task force of the American College of Neuropsychopharmacology carried out a review of the literature regarding potential associated factors that may influence remission in terms of timing and stability. Among others, these factors include the type of treatment, the dose, the treatment duration, the baseline severity of depressive symptoms, the stage
of treatment resistance (Treatment Resistant Depression [TRD] stage), compliance, the presence
of Diagnostic and Statistical Manual of Mental Disorders–Fourth Edition (DSM-IV) Axis I, II, or
III conditions, environmental stressors, the degree of social support, the retrospective morbidity of the illness, as well as neurobiological and genetic vulnerability.
There is no general consensus about the length of time the depressed patient in remission
should remain in remission for it to qualify as genuine, but according to most experts, this period of remission needs to last for between 4 and 6 months to be of clinical relevance to the patient. The length of the remission period is obviously of great clinical importance to the patient,
and it is also dependent on the continuation of antidepressant treatment.
Unfortunately, a significant number of patients do not achieve a fully symptom-free state, and
display residual subsyndromal depression or subthreshold depression. These patients have been
shown to have a higher risk of early relapse into depression, lower levels of social and psychological functioning, and greater rates of physical morbidity for conditions such as cardiovascular disease and stroke, as well as higher rates of mortality. Several therapeutic strategies have
been proposed to achieve remission or treat residual symptoms in patients suffering from major
depressive disorder. The most frequent residual symptoms targeted include anxiety, sleep disturbances, depressed mood, work difficulties, fatigue, and lack of interest. Among such residual depressive symptoms, severe chronic current insomnia—one of the most frequently observed manifestations of sleep disturbance in depression—appears to be an important residual core symptom
of depression, which may be related to the persistence of cognitive problems such as asthenia,
anhedonia, trouble in concentrating, and short-term memory difficulties. The rather high manifestation of residual symptoms observed in non fully remitted depressed patients justifies the need
for research into various therapeutic strategies such as switching, augmentation, and combination therapies, including with cognitive behavioral therapy, and the search for new targets to develop novel and more efficacious antidepressant treatments. There is a general consensus among
experts that full clinical remission after acute antidepressant treatment should be the gold standard and one of the priority objectives to be achieved in modern antidepressant therapy. 114 MEDICOGRAPHIA, VOL 31, No. 2, 2009
Defining remission in depression: the challenge of complete recovery – Mendlewicz
ÉDITORIAL
Définir la rémission dans la dépression :
le défi de la guérison complète
par J. Mendlewicz, Belgique
L
ES TROUBLES DÉPRESSIFS CONSTITUENT UN PROBLÈME DE
santé publique majeur, à tel point que l’Organisation Mondiale de
la Santé a estimé qu’en 2010 elle pourrait atteindre le 2 e rang parmi l’ensemble des maladies par ordre de prévalence, et ainsi contribuer de façon majeure à la charge pathologique globale chez l’homme. Les avancées du traitement des troubles dépressifs constitueront par conséquent un défi majeur que devront relever la
communauté médicale et la société dans son ensemble au cours des années à venir. Le concept
de rémission clinique dans le traitement de la dépression majeure a fait l’objet d’une attention
croissante au cours des dernières années. Les raisons de cet intérêt relativement récent sont
multiples : les patients déprimés ainsi que les organisations de patients ne sont pas totalement
satisfaits de l’efficacité et de la tolérance actuelles des antidépresseurs disponibles. Malgré les
bénéfices évidents apportés par les antidépresseurs, un grand nombre de patients déprimés qui
souffrent toujours de symptômes résiduels incapacitants sont exposés à un risque plus important de rechute ou de récidive par rapport à ceux ayant atteint une rémission complète après un
traitement antidépresseur. Les patients obtenant une rémission complète après traitement présentent un meilleur niveau de fonctionnement et un pronostic plus favorable par rapport aux
patients dont la guérison n’est pas complète.
Une rémission clinique adéquate est par conséquent d’une importance capitale pour le fonctionnement du patient, car il semble qu’elle constitue un facteur de prédiction de la stabilité à
long terme et un indicateur relativement satisfaisant d’un meilleur fonctionnement psychologique, ce dernier jouant un rôle de la plus haute importance pour l’évaluation de la qualité de
vie des patients déprimés. Pour toutes ces raisons, la communauté scientifique, mais également
les patients, se sont particulièrement intéressés à la prise en compte non seulement des taux de
réponse, mais également des taux de rémission, afin d’évaluer l’efficacité clinique réelle des antidépresseurs dans la pratique quotidienne et évaluer les nouveaux traitements au cours d’études
cliniques portant sur l’efficacité. Ce numéro de Medicographia rassemble les contributions d’experts dans le domaine des troubles de l’humeur (affectifs) qui aborderont sous des angles différents les problèmes essentiels liés au concept de rémission complète dans la dépression.
H. J. Möller et coll. examinent les connaissances actuelles sur l’évolution naturelle d’un épisode dépressif, les symptômes pouvant constituer des facteurs prédictifs de la réponse et d’une rémission, et l’évolution en fonction du temps de leur régression au cours d’un traitement antidépresseur.
D. Baldwin et A. Lopes s’intéressent plus particulièrement aux différents effets des antidépresseurs sur la dépression endogène et la dépression associée à des troubles anxieux, dans le cadre
de l’évolution de la dépression en fonction du temps et de la qualité de la rémission dans la dépression.
P. Monteleone et M. Maj décrivent l’importance des troubles des rythmes circadiens dans la dépression, et l’impact que la resynchronisation des rythmes circadiens peut avoir sur la qualité
de la rémission.
C. Soldatos et C. Theleritis se penchent sur la qualité du cycle sommeil-veille comme marqueur potentiel de la rémission dans la dépression. Ils abordent également un certain nombre de
problèmes comme la persistance des perturbations du sommeil après un traitement antidépresseur, et les perspectives ouvertes par les antidépresseurs ayant la capacité de rétablir le sommeil.
Définir la rémission dans la dépression : le défi de la guérison complète – Mendlewicz
MEDICOGRAPHIA, VOL 31, No. 2, 2009 115
ÉDITORIAL
S. Kennedy et S. Rizvi soulignent l’importance de maintenir un fonctionnement sexuel et
une qualité de vie satisfaisants chez les patients présentant une rémission à la suite d’une dépression, ce qui constitue également un facteur essentiel de l’observance du traitement.
J. Price et G. Goodwin analysent l’impact d’un traitement antidépresseur sur la réactivité cognitive et émotionnelle après une rémission, et les implications de celle-ci vis-à-vis des rechutes.
E. Paykel dresse la liste des symptômes résiduels dont la présence traduit une rémission incomplète et souligne la nécessité d’un traitement antidépresseur d’entretien, à laquelle une thérapie cognitive peut être associée, afin d’éviter le risque de rechute.
C. Muñoz dresse un tableau du mode d’action pharmacologique et des bénéfices cliniques de
l’agomélatine, un nouvel antidépresseur mélatoninergique.
W. Choucha et J. F. Allilaire discutent des différentes approches de la prise en charge clinique
à long terme pour le traitement des patients ayant présenté une rémission.
G. Fava et D. Visani préconisent l’utilisation séquentielle de la pharmacothérapie et de la
psychothérapie pour la prise en charge des symptômes résiduels chez les patients ayant guéri
d’une dépression, la considèrent comme le modèle le plus efficace pour éviter les rechutes et les
récidives et insistent sur les conséquences de cette méthode en ce qui concerne la programmation du traitement.
J. D. Guelfi réfléchit sur les mérites et les inconvénients des différentes méthodes d’évaluation
des rémissions, des rechutes et des symptômes résiduels dans la dépression.
La définition de la rémission est variable dans la littérature, où sont soulevées un certain
nombre de questions, notamment : la limite entre une rémission complète et une rémission partielle, la présence après le traitement de symptômes résiduels et le retour (ou non) au fonctionnement psychosocial prémorbide. Selon la conférence de consensus de la Fondation MacArthur,
la réponse clinique est définie comme une période au cours de laquelle est observée une certaine
amélioration des symptômes, mais d’amplitude insuffisante pour correspondre à l’obtention
d’une rémission complète, et s’accompagnant de la persistance de certains symptômes résiduels.
Sur les échelles d’évaluation de la dépression, cette situation correspond généralement à une
amélioration d’au moins 50 % des scores. En revanche, une rémission complète est obtenue
lorsque l’amélioration clinique est telle que le patient devient pratiquement asymptomatique.
La rémission clinique est généralement définie par un score inférieur ou égal à 7 sur l’Échelle
d’évaluation de la dépression de Hamilton en 17 items (17-item Hamilton Rating Scale for Depression, HAM-D17 ) ou un score inférieur ou égal à 10 à 12 sur l’Échelle d’évaluation de la dépression de Montgomery-Åsberg (Montgomery-Åsberg Depression Rating Scale, MADRS). La
persistance d’une rémission durable pendant l’administration d’un traitement antidépresseur
d’entretien revêt une importance clinique évidente, dans le cadre du concept de guérison.
La guérison clinique ne peut être établie chez un individu qu’après la persistance d’une rémission pendant au moins 3 ou 4 mois. Un groupe de travail du Collège américain de neuropsychopharmacologie (American College of Neuropsychopharmacology) a effectué un examen
de la littérature concernant les facteurs potentiellement associés susceptibles d’influencer la
rémission quant au moment de sa survenue et à sa stabilité. Ces facteurs comprennent entre
autres le type de traitement, la posologie, la durée du traitement, la sévérité initiale des symptômes dépressifs, le stade de résistance au traitement (stade de dépression résistant au traitement, Treatment Resistant Depression [TRD] stage), l’observance du traitement, la présence de
conditions correspondant à l’axe I, II ou III du Manuel Diagnostique et Statistique des Troubles
Mentaux 4e Édition (Diagnostic and Statistical Manual of Mental Disorders–Fourth Edition,
DSM-IV), les facteurs de stress environnemental, le degré de soutien social, la morbidité rétrospective de la maladie, ainsi que la vulnérabilité neurobiologique et génétique.
Il n’existe pas de consensus global sur la durée permettant de qualifier une rémission de
réelle, mais, selon la plupart des experts, cette période doit être au moins de 4 à 6 mois pour
avoir une signification clinique pour le patient. La durée de la période de rémission est naturellement d’une importance clinique extrême pour le patient, et est conditionnée par la poursuite du traitement antidépresseur.
Malheureusement, un nombre significatif de patients n’atteignent jamais un état asymptomatique, et continuent pendant un certain temps à présenter une dépression sous-syndromique
résiduelle ou une dépression infraliminaire. Il est démontré que ces patients présentaient un
risque aggravé de rechute dépressive précoce, des niveaux inférieurs de fonctionnement social
et psychologique, et des taux supérieurs de morbidité physique à type de maladies cardio-vasculaires et d’accidents vasculaires cérébraux, ainsi que des taux de mortalité plus élevés. Plu116 MEDICOGRAPHIA, VOL 31, No. 2, 2009
Définir la rémission dans la dépression : le défi de la guérison complète – Mendlewicz
ÉDITORIAL
sieurs stratégies thérapeutiques ont été proposées pour atteindre la rémission ou traiter les
symptômes résiduels chez les patients souffrant de dépression majeure. Les symptômes résiduels plus fréquents visés par les traitements comprennent : l’anxiété, les troubles du sommeil,
l’humeur dépressive, les difficultés professionnelles, la fatigue et la perte d’intérêt. Parmi ces
symptômes dépressifs résiduels, l’insomnie chronique sévère – l’une des manifestations les plus
fréquemment observées de troubles du sommeil dans la dépression – semble constituer un symptôme central résiduel important de la dépression, qui pourrait être lié à la persistance de problèmes cognitifs, notamment l’asthénie, l’anhédonie, les troubles de la concentration et les
difficultés de mémorisation à court terme. La fréquence relativement élevée des symptômes résiduels observés chez les patients déprimés n’ayant pas obtenu une rémission complète justifie
la nécessité de rechercher différentes stratégies thérapeutiques, notamment des traitements de
substitution, adjuvants et d’association, en particulier avec une thérapie comportementale cognitive, et l’identification de nouvelles cibles pour développer des traitements antidépresseurs
nouveaux et plus efficaces. Les experts sont arrivés à un consensus sur le fait que la rémission
clinique complète après un traitement antidépresseur aigu doit être la référence et l’un des objectifs prioritaires à atteindre dans le traitement moderne de la dépression. Définir la rémission dans la dépression : le défi de la guérison complète – Mendlewicz
MEDICOGRAPHIA, VOL 31, No. 2, 2009 117
IN
DEPRESSION: THE ULTIMATE GOAL
TRUE REMISSION
Hans-Jürgen MÖLLER, MD, Prof
Florian H. SEEMÜLLER, MD
Michael RIEDEL, MD, Prof
Department of Psychiatry
Ludwig-Maximilians University
Munich, GERMANY
Time course of response
and remission during
antidepressant treatment
b y H . - J . M ö l l e r , F. H . S e e m ü l l e r ,
and M. Riedel, Germany
ajor depression is a complex and inhomogeneous illness with an etiopathogenesis that is based upon multiple factors that may act at different levels (psychological, biological, genetic, and social). It is a
matter of fact that one single treatment, or one single treatment approach, can
therefore never be adequate for such a diverse illness. There is hope that in the
near future we will be able to treat patients on an even more individual level
by using genetic or brain morphologic markers. In the case of drug resistance,
pseudoresistance—with special attention given to adherence—should be ruled
out first. Response to treatment can occur very early, even within the first 2
weeks of the treatment course. In clinical trials, the highest response rates appear within the first 4 weeks. When, exactly, and to what extent response appears, also depends on its exact definition. Two definitions have achieved acceptance in the research community—that of early improvement (20% reduction
on the Hamilton Rating Scale for Depression [HAM-D] within the first 2 weeks)
and that of response (50% reduction of the initial HAM-D score). The first
may be especially useful in monitoring early symptom alleviation due to the
respective treatment, while the second may be more useful in detecting sustained changes, especially when combined with a time criterion. Both are highly predictive of remission, which is usually observed at a lower rate compared
with response, but which in clinical trials surpasses the response rate from week
4 to 6 onward. Thus, response to treatment may be considered as the precursor of remission, ie, the phase inaugurating the natural healing process induced by antidepressant therapy and which, in the majority of cases achieves
its goal, ie, remission, the complete relief of all symptoms.
M
Medicographia. 2009;31:118-125.
echanisms involved in the emergence of
depressive episodes are multifactorial and
not yet fully understood. As recently pointed out by Kenneth Kendler, a comprehensive etiological understanding of psychiatric disorders will
require the integration of multiple explanatory perspectives.1 If we want to shed light on the complex
interactions between the pathophysiology of depression, its treatment, and its cure, we need to consider all the different psychopathological subtypes,
the contribution of different neurobiological and
psychosocial etiopathogenetic factors, the clinical
response to treatment, and the long-term prognosis, and additionally the comorbidity with other psychiatric or somatic disorders. There is enormous
variation between individual patients with regard to
these different factors, which corresponds to the
variability of outcome regarding the time course
of response and remission. In the following paragraphs, we will first try to summarize some of the
most recent findings concerning the impact on de-
M
(see French abstract on page 125)
Keywords: major depression; response; remission; time course; etiopathogenesis
www.medicographia.com
Address for correspondence: Prof Dr med Hans-Jürgen Möller, Department of Psychiatry and
Psychotherapy, Ludwig-Maximilians University Munich, Nussbaumstrasse 7, 80336 Munich, Germany
(e-mail: [email protected])
118 MEDICOGRAPHIA, VOL 31, No. 2, 2009
Natural evolution
of a depressive episode
SELECTED
ABBREVIATIONS AND ACRONYMS
BDNF
CRH
ECT
HAM-D
MADRS
brain derived neurotrophic factor
corticotropin releasing hormone
electroconvulsive therapy
Hamilton Rating Scale for Depression
Montgomery–Åsberg Depression Rating
Scale
MDD
major depressive disorder
SSRI
selective serotonin reuptake inhibitor
STAR*D Sequenced Treatment Alternatives to
Relieve Depression (study)
Time course of response and remission during antidepressant treatment – Möller and others
TRUE REMISSION
Stressful life events and gene-environment
interaction
Stressful life events that occur during an individual’s lifespan influence the onset and course of major depression. In a recent paper by Kendler and
colleagues,3 the relationship between dependent
stressful life events (life events that result from the
respondents’ own behavior) or independent stressful life events (events that mostly result from “bad
luck”) and the onset of major depression was systematically investigated in a sample of 1898 female
twins (monozygotic and dizygotic).3 Independent
and dependent stressful life events were associated
to a highly significant extent with new onset of depressive episodes, and the strongest association was
observed for the dependent events. The odds for the
onset of major depression in the month of a life
event were 5.64 for all subjects.3 However, major depression can also arise without any prior life event
and the vast majority of people experiencing life
events do not develop depression. One reason for
this might be that the impact of life on the development of an illness may be moderated by a geneenvironment interaction. In 2003, Avshalom Caspi
prospectively investigated why life events lead to
depression in some people but not in others.4 In a
representative birth cohort of 1037 children who
had had comprehensive psychiatric assessments at
the ages of 3, 5, 7, 9, 11, 13, 15, 18, and 21 years, a
functional polymorphism of the promoter region
of the serotonin transporter gene (5-HTT) was found
to moderate the influence of stressful life events on
depression.4 Specifically, the study revealed three
major results. First, with regard to the gene-environment interaction, it was found that life events
after the 21st birthday significantly predicted a new
onset of depression at age 26 among carriers of an
S-allele who had no history of prior depression, but
did not predict onset of depression among l/l homozygotes. Second, even suicide ideation, which is always susceptible to having an underlying biological mechanism, could be significantly predicted for
individuals with the S-allele but not for l/l homozygotes (Figure 1). Third, childhood maltreatment
during the first life decade also significantly predicted adult depression among S-allele carriers but
not among l/l homozygotes. In addition, the sole relationship between genotype and depression not
associated with a life event was not significant.4 In
summary, this model further supports the concept
that genetic variants with a high prevalence in the
general population probably act to “promote an organism’s resistance to environmental pathogens.” 4
Monoamine deficiency hypothesis
On a transmitter level, the monoamine deficiency
hypothesis remains of extreme importance. Apart
from the fact that all antidepressant agents have an
DEPRESSION: T
HE
ULTIMATE GOAL
effect on at least one of the monoamine transmitters (dopamine, serotonin, norepinephrine), there
is also compelling evidence from the opposite direction. Tryptophan is rate limiting for serotonin
synthesis in the brain. Oral tryptophan depletion
does not induce depression in mentally healthy subjects, but will cause depression in patients successfully treated with a selective serotonin reuptake
inhibitor (SSRI). Likewise, α-methyl paratyrosine
does not induce depression in normal people, but
does in patients who have been treated with a norepinephrine reuptake inhibitor.5 The importance of
dopamine in depression is highlighted by the high
comorbidity rates of depression in patients with
Parkinson’s disease, the antidepressant effect of buproprione, and the depressogenic effect of dopamine
depletion with reserpine.
0.16
S/S
0.14
Probability of suicide
ideation/attempt
pression of four major influential levels; namely,
the psychological, environmental, biological, and
genetic levels. An excellent overview of the most recent concepts in our understanding of the biological mechanisms of major depression can be found
in reference 2.
IN
0.12
0.10
S/l
0.08
0.06
l/l
0.04
0.02
0.00
0
1
2
3
Number of stressful life events
4+
Figure 1. Relationship between suicidal ideation and attempts, number
of stressful life events, and 5-HTT allele carrier status.
After reference 4: Caspi A, Sugden K, Moffitt TE, et al. Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science. 2003;301:386-389.
Copyright © 2003, The American Association for the Advancement of Science.
Hypothalamic-pituitary-adrenal axis
Another possible link between life stress and depression might be the moderation of depression
through the hypothalamic-pituitary-adrenal axis.
Depressed subjects can show elevated plasma cortisol levels6 and elevated corticotropin releasing
hormone (CRH) levels in the cerebrospinal fluid.7
Furthermore, the normal cortisol suppression response is absent in about half of the most severely
depressed subjects.8 Consequently, antidepressantinduced remission is also associated with the reversal of the abnormal suppression test, supporting
the notion of a causal relationship.8 Most recent research for new antidepressant agents has also focused on glucocorticoid receptor blockers, with
promising preliminary results.9
Higher glucocorticoid levels may also be associated with reduced neurogenesis, and have been
linked to a decreased hippocampal size in patients
with depression.10 Brain derived neurotrophic factor (BDNF), which plays an important role in neurogenesis involving neuronal plasticity, synaptic
growth, and neuronal cell survival, was found to be
significantly reduced in a cohort of suicide completers.11 Antidepressant treatment options such as
Time course of response and remission during antidepressant treatment – Möller and others
MEDICOGRAPHIA, VOL 31, No. 2, 2009 119
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
regular physical activity,12 electroconvulsive therapy (ECT)13 or antidepressant compounds14 have
been shown to significantly elevate BDNF levels.
Circadian rhythms
Circadian disturbances occur in depression and include not only disturbances to physiological parameters (eg, body temperature) or biological parameters such as cortisol secretion, but also mood and
the sleep-wake cycle. Therapeutic treatments aimed
at the regulation of these disturbances in depressed
patients have been shown to have a significant impact in relieving symptoms of depression.15
It is not surprising given this diversity and complexity in the etiopathogenesis of depression and the
pharmacological mechanisms of antidepressants
that there is a broad range of possible answers to
the question of which therapeutic approach is the
most beneficial for the individual patient. For such
a highly inhomogeneous and diverse medical condition, response and remission can hardly be achieved
with only a single therapeutic strategy. Today, we
still need to make use of the full range, variety, and
combination of different therapeutic strategies, such
as inpatient and outpatient treatment, the differential and sequential use of psychopharmacological
compounds, pharmacological combination and
augmentation strategies, as well as other highly effective biological treatments including light therapy, ECT and transcranial magnetic stimulation, and
psychotherapy, in order to deal with this illness according to the rules of good clinical practice.16 Apart
from subtyping depression and using predictors,
which will be discussed later, good allies in that
challenge remain time and patience to obtain the
optimal treatment regimen for each individual patient. This notion finds support in naturalistic inpatient data from a large German inpatient study, as
well as in the outpatient Sequenced Treatment Alternatives to Relieve Depression (STAR*D) study.17,18
In both trials, the patients were actively involved in
the decision-making process of their next treatment
step, and results of both trials indicate that the
longer the treatment and thus the time to individualize and optimize the treatment regimen, the better are the response and remission rates.17,18
Evolvement of a major depressive episode:
the psychopathological perspective
The natural evolution and time course over which
depressive symptoms usually first develop has rarely
been systematically investigated. However, better
knowledge about early warning signs and the development of major depressive episodes could significantly improve health education and might lead
to an improvement in early detection.
One of the most consistent early warning signs
and prodromal symptoms of major depression is
sleep disturbance.19-21 Sleep serves as an important
timer for the daily circadian rhythm and is found to
be disturbed about 5 weeks prior to the new onset of
a depressive episode in patients with recurrent depression.20 Data from the National Institute of Mental Health catchment area program across four sites
120 MEDICOGRAPHIA, VOL 31, No. 2, 2009
in the US found strong associations between sleep
disturbances and the depressive symptoms of diminished sexual drive, feelings of worthlessness or
excessive guilt, and trouble concentrating or thinking, 1 year prior to the new onset of a major depressive episode.21,22 In very fast developing episodes, loss
of libido and suicidality can be the first symptoms
to develop. This is of special importance, because
loss of libido often tends to be overlooked clinically, since clinicians are often reluctant to ask questions about a patient’s sexual life. In addition, these
results do suggest that suicidality should be given
notably more attention in patients with a fast onset
of a depressive episode.23 The core and illness-defining symptoms that develop in the further course of
depression are depressed mood and/or the inability to experience joy (anhedonia).
Different depressive syndromes can develop, depending on which symptoms evolve. From a psychopathological perspective, the most important
ones that we differentiate today, because of their
different prognostic and therapeutic implications,
are melancholic, atypical, and psychotic depression.
Recently, Gin Mahli and Gordon Parker developed
a model for the relationship between psychotic
melancholia, nonpsychotic melancholia, and nonmelancholic depression, and the major monoamines
thought to be involved in depression. The authors
postulate that in nonmelancholic depression with
its primary affective symptoms, serotonergic dysfunction may prevail, while nonpsychotic melancholia, which is accompanied more by psychomotor disturbance, derives from a more pronounced
norepinephrine dysfunction. With respect to psychotic depression, dopamine may be involved to a
higher degree (Figure 2).24 As another example, the
subtype of atypical depression can be mentioned
here, which has long been seen as an indication for
treatment with monoamine oxidase inhibitors.25,26
Finally, in a full-blown depressive episode, depressive symptoms appear in the following percentages (data from 1014 depressed inpatients using
the 21-item Hamilton Rating Scale for Depression
[HAM-D21]; given in declining order of percentage):
depressed mood 99.4%, loss of interest in work and
activities 99.2%, somatic symptoms 85%, psychic
anxiety 84.3%, feelings of guilt 78.5%, insomnia
middle of night 78.3%, genital symptoms 76.6%,
suicidal ideation 76.5%, insomnia early night 71.4%,
somatic anxiety 71.2%, insomnia late night 66.8%,
diurnal variation 66.5%, loss of appetite 64.4%, psychomotor retardation 55.6%, agitation 49%, loss of
weight 48.6%, hypochondriasis 39%, depersonalization 30.9%, paranoid ideas 21.5%, lack of illness insight 18%, and compulsiveness 10.1%.27 The distinct pattern or cluster of symptoms that comes to
the fore in an individual could lead the way toward
finding a more individualized treatment regimen,
which might speed up the time to response. For
example, in patients with anxiety symptoms, a combination of an antidepressant with a benzodiazepine
may lead to a quick reduction in the sensation of
fear. Also, sleep disturbances can be addressed specifically with hypnotic comedication or a sedating antidepressant.
Time course of response and remission during antidepressant treatment – Möller and others
TRUE REMISSION
DEPRESSION: T
IN
Depressive
subtype
Clinical
feature
Neurotransmitter
Psychotic
melancholia
Psychotic
features
DA
Nonpsychotic
melancholia
Psychomotor
disturbance
NE
Nonmelancholic
depression
Mood
disorder
5-HT
Structural model
Despite the fact that response and remission are
gaining more and more acceptance in the research
community as major outcome criteria in clinical
trials as a result of their important clinical implications compared with mean-score differences, most
trials still rely solely on end point differences between drug and placebo for remission or response
rates. Only a few report both rates, and even fewer
also report the time course of response and remission rates.
Generally speaking, the course of remission and
response under treatment seems to follow a typical
pattern: the strongest effects in terms of a mean
score reduction usually appear at the beginning of
the antidepressant therapy regimen, not only in
phase 3 studies sponsored by pharmaceutical companies but also in naturalistic studies. By far the
most pronounced HAM-D mean score decline can
thus be seen after the first 2 weeks of a newly initiated treatment. Thus in the aforementioned naturalistic trial involving 1014 inpatients, a 34% reduction in the HAM-D21 score and a 33% reduction in
the Montgomery–Åsberg Depression Rating Scale
(MADRS) score was evident after week 2.17 This is in
excellent accordance with data from another naturalistic trial showing a 30% MADRS reduction after 2 weeks in 11 760 outpatients with major depressive disorder (MDD) treated with escitalopram.28
Clearly, nonspecific factors such as the inpatient
setting or professional attention received by patients
within clinical trials, and also fast-acting comedication like benzodiazepines, contribute to such fast
symptom relief and support the intrinsic effect of
antidepressants.
ULTIMATE GOAL
Figure 2. Functional and
structural model of different depressive subtypes.
Abbreviations: DA, dopamine;
NE, norepinephrine; 5-HT,
5-hydroxy-trytophan/serotonin.
After reference 24: Malhi GS,
Parker GB, Greenwood J.
Structural and functional models of depression: from subtypes
to substrates. Acta Psychiatr
Scand. 2005;111:94-105.
Copyright © 2004, Blackwell
Munksgaard.
Functional model
end to about 7%.18 Remission rates started at about
11% after week 2 and went up to about 22% at week
6 and declined again to 14% at study end. Interestingly, until week 4, there were higher response than
remission rates, and from week 6 onward, the relationship was the opposite, with higher remission
than response rates (Figure 3).18 With respect to the
latter phenomenon, a strikingly similar picture is
revealed by our own data on naturalistic-treated inpatients (Figure 4, page 122).17,18 At week 6, the remission rate surpasses the response rate up to the
final visit. This clearly justifies and underlines the
importance of differentiating and calculating both
response and remission rates as two different outcome variables. These results further imply that response usually precedes remission, which is also
strengthened by the notion that achievement of response is highly predictive of remission.
Achievement of remission is highly predictive of
good functional recovery and lower relapse rates.
Thus responding to an antidepressant therapy might
represent advancement of the healing process in
25%
Remission (n=943)
Response (n=1343)
20%
Percentage of patients
What symptoms are predictive
of remission and what is the timing
of their alleviation?
HE
15%
10%
5%
0%
2
4
6
8
10
12
13
Week of treatment
Time course of response versus remission
The time courses of remission rates versus response
rates reveal a contrasting picture. In the first step
of STAR*D, an investigation of citalopram over 14
weeks in 2876 outpatients with depression, the response rate was approximately 17% at week 2; subsequently, the highest response rate was at week 4
with 25%, with a constant decline thereafter to study
Response was defined as improvement of 50% in QUID-SR score from baseline.
Remission was defined as a QUID-SR score of 5 at end point.
Figure 3. Remission and response rates from Sequenced Treatment Alternatives to
Relieve Depression (STAR*D) Level 1. QUID-SR, Quick Inventory of Depressive
Symptomatology—Clinician Rated.
After reference 18: Trivedi MH, Rush AJ, Wisniewski SR, et al. Evaluation of outcomes with citalopram
for depression using measurement-based care in STAR*D: implications for clinical practice. Am J Psychiatry. 2006;163:28-40. Copyright © 2006, American Psychiatric Association.
Time course of response and remission during antidepressant treatment – Möller and others
MEDICOGRAPHIA, VOL 31, No. 2, 2009 121
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
30%
Percentage of patients
25%
Remission (n=465)
Response (n=498)
20%
15%
10%
5%
0%
2
4
6
8
10
Later
Week of treatment
Figure 4. Remission and
response rates from a
German naturalistic trial.
Based on data from
reference 17.
major depression. Stassen and coworkers analyzed
data from 2848 patients included in double-blind
randomized controlled trials, and found no difference between active drug and placebo regarding the
time of onset of improvement, which was equally
distributed in the placebo and active drug groups.
However, significant interindividual differences in
response patterns independent of the treatment
condition occurred. In addition, significant differences between the active group and the placebo
group could be seen in the total percentage of improvers. The authors conclude that affectively ill
patients are likely to possess a common biological
“resilience-like component” that largely controls
recovery and can best be studied in placebo-treated
patients. Active drugs might induce this natural resilience mechanism.29
Timing of onset of antidepressant action
Traditionally, it has been thought that standard antidepressants take about 1 month for their action
to fully unfold, and that they have a delayed onset
of action of at least 2 weeks. Identification of the exact timing of the onset of antidepressant action has
been a well-debated matter for the last 20 years.
Originally, Frederic Quitkin proposed in his pattern
analytic approach that real drug-placebo differences
could not be observed before 3 weeks of treatment.30
An earlier improvement was associated with a placebo response and a lack of sustained improvement,
whereas the opposite was true for true drug responders, who showed a delayed but sustained onset of
improvement.30,31 More recently, this view has been
questioned by a large number of authors, including
Stassen and Szegedi, who have not only emphasized
that an earlier onset of improvement before 2 weeks
was highly prevalent, but also showed that it was
highly predictive of later outcome.15,32-34 An early
improvement was therefore defined as a 20% reduction in the initial HAM-D17 score within the first
2 weeks. Today, there is strong evidence that early
onset of antidepressant action exists and appears to
be highly sensitive for later response or remission.
These results can best be translated into clinical
122 MEDICOGRAPHIA, VOL 31, No. 2, 2009
practice as follows: if after 2 weeks no improvement
at all can be observed under a new antidepressant
regimen, then the pharmacologic regimen should
be adjusted or changed early rather than waiting
for another 2 or 3 weeks. In this case, if no alteration of the treatment has occurred, the chances of
this person still responding are below 20%.34
However, there are a number of methodological
pitfalls inherent in the study of this phenomenon.
The rating scales currently in use such as HAM-D
and MADRS were never developed to detect early
changes in psychopathology due to antidepressant
action. In addition, most publications on this topic rely on post-hoc analysis of phase 3 trials not designed for the detection of an early onset of action,
with 2-week rating intervals not allowing the detection of earlier improvements. The inconsistencies in Quitkin’s conclusions can best be explained
by methodological differences between the two approaches. Quitkin et al defined stable drug response
very strictly as “much improvement” on the clinical global impression scale. As pointed out by Gelenberg, it might be that the lag in the onset of antidepressant action observed by Quitkin was caused
by the high degree of improvement necessary for response.10 In other words, authors such as Stassen
and Szegedi observed earlier effects, because they
used lower and more sensitive cut-off values, which
enabled them to detect minor changes earlier.
One of the few studies specifically designed to
study the onset of action of antidepressants was
conducted by Rojo and coworkers. They investigated the onset of antidepressant action in 582 depressed outpatients with HAM-D17 assessments at
baseline and after 1, 2, and 4 weeks.35 Patients were
treated with mirtazapine and, depending on the
timing of response, sustained responders were subdivided into four groups: very fast responders, fast
responders, traditional responders, and nonresponders. At baseline, only a few symptoms captured by
HAM-D17 showed significant differences between the
responder groups; ie, items such as guilt, suicide,
and genital symptoms: the more severe these baseline variables were, the slower was the response.36
Order and timing of symptom alleviation
The order and timing of the alleviation of depressive symptoms has also rarely been studied. With respect to the Rojo study, across all four subgroups,
the pattern on a symptom level did not vary: first, all
groups improved on items such as depressed mood,
early insomnia, and psychic anxiety. Second was
improvement on the HAM-D item “general somatic symptoms.” Interestingly, this improvement pattern occurred for the very fast group within the first
week, for the fast group within the second week, and
for the traditional group within the third week. In
summary, the alleviation of HAM-D17 symptoms for
each subtype was similar, but occurred at a different time in each responder group.36 These results
suggest that this equal response may not depend
on personality traits, doctor-patient relationships,
or medical practice, but rather on pharmacological factors and pharmacokinetics, at least in the case
of mirtazapine.
Time course of response and remission during antidepressant treatment – Möller and others
TRUE REMISSION
Clinical predictors of response to antidepressant
medication
One way to classify predictors is to distinguish between biological and clinical predictors, meaning
either predictors that can be measured with a distinct laboratory test or predictors that can simply
be observed or asked about. In clinical research, in
addition to the classification of subtypes of depression, clinical/anamnestic predictors have been examined regarding the response to antidepressants.
The following have been relatively consistently described as being relevant to a rather poor response
to antidepressants37,38 — most have been also confirmed in later examinations18,39,40: poor social adaptation, neurotic traits in the premorbid personality,
number and duration of earlier psychiatric inpatient
treatments, nonresponse to earlier treatments with
antidepressants, chronicity of the depressive symptoms, mild degree of depressive symptoms, delusions, and absence of vital symptoms.
However, the variance rates described by single
predictors are for the most part so minor that they
are hardly useable for practicable prognostics in individuals. In addition, there are hardly any hypotheses-free approaches investigating a whole set
of different psychopathologic measures and clinical
variables with respect to their prognostic meaning.
So far, the predictors we know of can at best con-
DEPRESSION: T
HE
ULTIMATE GOAL
tribute to group statistical differentiation. Future
research should also focus on the possibility of combining predictors, thereby potentially enabling prognosis to be optimized.
Why are available antidepressants
not always able to achieve remission?
Partial response and nonresponse to antidepressant
medications are a common problem in patients with
depression. Between 10% and 30% of depressed patients taking an antidepressant are partially or totally resistant to the treatment. Recurrence of depression while still taking medication (ie, breakthrough)
can also occur. In our own cohort of 1014 depressed
inpatients, about 32% of all patients suffered from
an index episode that lasted longer then 6 months
and were therefore classified as partially refractory
patients.17
All patients
4
Item
Item
Item
Item
Item
Item
3
Score
In our own naturalistic follow-up study on 1014
depressed inpatients, we observed the long-term alleviation of the 6 core items of HAM-D (Seemüller
et al, unpublished data). Apart from item 1, all of
the other 5 items6,10,11,23,35 paralleled the same decline pattern until discharge, with the most pronounced difference between baseline and week 2.
Item 1 of HAM-D17 , however, referred to as “depressive feelings,” showed the strongest and fastest
decline of all items up to discharge (Figure 5). In
another large naturalistic trial on escitalopram in
11 760 outpatients, patients were asked to name the
MADRS symptoms that changed the most 2 weeks
after starting antidepressant therapy. Patients most
frequently answered that “inner tension,” followed
by “sadness,” “pessimistic thoughts,” and “reduced
sleep” had changed as early as week 2.28
Unfortunately, a substantial proportion of patients do not respond to treatment within this short
time period, and most depressed patients do not
initially achieve remission. In STAR*D, only 28%
of the initial sample of 2876 depressed patients
achieved remission, and 47% were responders with
citalopram.18 The longer the search for an effective
antidepressant treatment in an individual, the higher the potential risk of chronicity, the higher the
overall costs, and the worse the functional outcome.
If a patient could be detected and identified early on
as being a nonresponder to an antidepressant and
could consequently be referred earlier to a more effective strategy such as dual-acting antidepressants,
augmentation strategies like lithium augmentation
or augmentation with an atypical antipsychotic, or
to other highly effective treatments such as ECT, the
difficult challenge of optimizing the treatment of
depression could be facilitated.
IN
1 (depressive feelings)
2
7
8
10
13
2
1
0
0
2
4
6
8
10
Week
When speaking of resistance, we should clearly
separate true drug resistance from what we call
“pseudoresistance.” Often, patients who are labeled
“treatment resistant” have not had a single adequate
medication trial (either in terms of dose or duration). Another often overlooked reason for pseudoresistance is misdiagnosis, and with it goes the tendency for many psychiatrists to stick to their original
diagnosis instead of reevaluating symptoms carefully. One of the most important reasons for pseudoresistance might be poor adherence. Particularly in outpatients, adherence rates can be as low as
50%, with the other 50% of patients not taking their
medication properly.41 In most cases, side effects account for the poor compliance rates. Olin and colleagues summarized the results of a survey of 1066
psychiatrists conducted at the 2007 American Psychiatric Association meeting. According to the survey, about one third of the respondents considered
current antidepressants to be only “somewhat effective,” and ranked sexual dysfunction, weight gain,
and inadequate efficacy as the primary roadblocks
to successful treatment.
Time course of response and remission during antidepressant treatment – Möller and others
Figure 5. Individual item
changes for the 6 core
symptoms on the 17-item
Hamilton Rating Scale
for Depression in naturalistic-treated depressed
patients.
MEDICOGRAPHIA, VOL 31, No. 2, 2009 123
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
In addition, comorbidities such as substance
abuse, personality disorders, and general medical
disorders including hypothyroidism and anemia can
also influence the overall success of a treatment
plan and can muddy the water. In particular, medical comorbidities might negatively influence outcome in two ways: first, through the direct impact
of the medical comorbidity on the psychiatric illness, and second, through adverse effects of medications used to treat the medical comorbidities, also
known as pharmacogenic depression. With regard
to relapse rates after complete remission, Andrew
Nierenberg demonstrated most recently in his talk
at the Collegium Internationale Neuro-Psychopharmacologicum congress in Munich, showing data
from STAR*D, that the higher the load for psychiatric comorbidities, the higher the risk of relapse
over 1 year.42
The reasons for true drug resistance remain unknown. Unfortunately, even the definition of drug
resistance is not fully standardized and varies greatly across different studies. In daily clinical practice,
probably the most useful definition might be as follows: drug nonresponse to at least two adequate
trials of antidepressants (adequate dosage and treatment duration) with different mechanisms of action. Given the aforementioned complexity and
diversity of the etiopathogenesis of such a multifaceted illness like depression, a “one treatment fits
all” approach to regaining a new homeostasis on a
transmitter level and a functional level at the same
time cannot be very fruitful.
New ideas regarding the underlying pharmacological mechanisms that cause a poor response may
lead to a more personalized therapy that is based on
“the right drug for the right patient.” One new way
forward might arise from a recent study on the
ABCB1 drug transporter gene, in which the basic
idea was that every antidepressant drug must pass
the blood brain barrier before its antidepressant acREFERENCES
1. Kendler KS. Explanatory models for psychiatric illness. Am J
Psychiatry. 2008;165:695-702.
2. Belmaker RH, Agam G. Major depressive disorder. N Engl J
Med. 2008;358:55-68.
3. Kendler KS, Karkowski LM, Prescott CA. Causal relationship
between stressful life events and the onset of major depression.
Am J Psychiatry. 1999;156:837-841.
4. Caspi A, Sugden K, Moffitt TE, et al. Influence of life stress on
depression: moderation by a polymorphism in the 5-HTT gene.
Science. 2003;301:386-389.
5. Ruhe HG, Mason NS, Schene AH. Mood is indirectly related
to serotonin, norepinephrine and dopamine levels in humans:
a meta-analysis of monoamine depletion studies. Mol Psychiatry.
2007;12:331-359.
6. Burke HM, Davis MC, Otte C, Mohr DC. Depression and cortisol responses to psychological stress: a meta-analysis. Psychoneuroendocrinology. 2005;30:846-856.
7. Merali Z, Du L, Hrdina P, et al. Dysregulation in the suicide
brain: mRNA expression of corticotropin-releasing hormone receptors and GABA(A) receptor subunits in frontal cortical brain
region. J Neurosci. 2004;24:1478-1485.
8. Carroll BJ, Cassidy F, Naftolowitz D, et al. Pathophysiology of
hypercortisolism in depression. Acta Psychiatr Scand. 2007(suppl);90-103.
9. Flores BH, Kenna H, Keller J, Solvason HB, Schatzberg AF.
Clinical and biological effects of mifepristone treatment for psychotic depression. Neuropsychopharmacology. 2006;31:628-636.
10. Frodl T, Schule C, Schmitt G, et al. Association of the brainderived neurotrophic factor Val66Met polymorphism with reduced hippocampal volumes in major depression. Arch Gen Psy-
124 MEDICOGRAPHIA, VOL 31, No. 2, 2009
tivity can unfold. P-glycoprotein (P-gp),43 which is
encoded on the ABCB1 gene, serves as a transporter
and acts as an efflux pump for a wide range of psychopharmacological compounds. Uhr and colleagues
were able to identify the antidepressant substrates
of P-gp, and could predict response and remission
in relation to the genotype of subjects who were
treated with antidepressants that were substrates
of P-gp.43 A completely different approach may arise
from the use of brain imaging techniques. Frodl and
coworkers, for example, were able to demonstrate
that nonresponders to antidepressant treatment
showed significantly lower amygdala volumes at
baseline than did remitters.35
Conclusion
In summary, aside from the complexity of this inhomogeneous illness, several factors may contribute to treatment failure, including undiagnosed or
misdiagnosed medical conditions such as hypothyroidism and anemia. The patient who does not respond or only partially responds to an antidepressant should therefore first be reassessed to make
sure the original diagnosis of depression was correct. Also, nonpsychiatric drugs such as methyldopa,
β-blockers, and reserpine can cause or exacerbate
depression, and comorbid disorders (eg, eating disorders, substance abuse or dependence) may affect
treatment response. Psychotic depression, melancholic depression, and atypical depression are depressive subtypes that may require concurrent
pharmacotherapy such as antipsychotic or augmentative psychotherapy. Finally, adverse effects and
poor compliance may be additional obstacles to successful treatment, but with our growing understanding of the pathophysiology of the illness, there
is hope that in the near future we will be making
progress in the direction of individualizing the clinical decision-making process. chiatry. 2007;64:410-416.
11. Karege F, Vaudan G, Schwald M, Perroud N, La Harpe R. Neurotrophin levels in postmortem brains of suicide victims and the
effects of antemortem diagnosis and psychotropic drugs. Brain
Res Mol Brain Res. 2005;136:29-37.
12. Tang SW, Chu E, Hui T, Helmeste D, Law C. Influence of exercise on serum brain-derived neurotrophic factor concentrations
in healthy human subjects. Neurosci Lett. 2008;431:62-65.
13. Marano CM, Phatak P, Vemulapalli UR, et al. Increased plasma concentration of brain-derived neurotrophic factor with electroconvulsive therapy: a pilot study in patients with major depression. J Clin Psychiatry. 2007;68:512-517.
14. Hellweg R, Ziegenhorn A, Heuser I, Deuschle M. Serum concentrations of nerve growth factor and brain-derived neurotrophic factor in depressed patients before and after antidepressant
treatment. Pharmacopsychiatry. 2008;41:66-71.
15. Kennedy SH. Agomelatine: an antidepressant with a novel
mechanism of action. Future Neurology. 2007;2:145-151.
16. Nierenberg AA, McLean NE, Alpert JE, Worthington JJ, Rosenbaum JF, Fava M. Early nonresponse to fluoxetine as a predictor
of poor 8-week outcome. Am J Psychiatry. 1995;152:1500-1503.
17. Seemuller F, Riedel M, Obermeier M, et al. Outcomes of 1014
naturalistic treated inpatients with major depressive episode.
Poster presented at: XXVI CINP Congress; July 13-17, 2008; Munich, Germany.
18. Trivedi MH, Rush AJ, Wisniewski SR, et al. Evaluation of outcomes with citalopram for depression using measurement-based
care in STAR*D: implications for clinical practice. Am J Psychiatry. 2006;163:28-40.
19. Hagerty BM, Williams RA, Liken M. Prodromal symptoms of
Time course of response and remission during antidepressant treatment – Möller and others
TRUE REMISSION
recurrent major depressive episodes: a qualitative analysis. Am J
Orthopsychiatry. 1997;67:308-314.
20. Perlis ML, Giles DE, Buysse DJ, Tu X, Kupfer DJ. Self-reported sleep disturbance as a prodromal symptom in recurrent depression. J Affect Disord. 1997;42:209-212.
21. Roper PD. Prodromal symptoms. Am J Psychiatry. 1989;146:
941-942.
22. Dryman A, Eaton WW. Affective symptoms associated with
the onset of major depression in the community: findings from
the US National Institute of Mental Health Epidemiologic Catchment Area Program. Acta Psychiatr Scand. 1991;84:1-5.
23. Hegerl U, Bottner AC, Holtschmidt-Taschner B, et al. Onset
of depressive episodes is faster in patients with bipolar versus
unipolar depressive disorder: evidence from a retrospective comparative study. J Clin Psychiatry. 2008;69:1075-1080.
24. Malhi GS, Parker GB, Greenwood J. Structural and functional models of depression: from subtypes to substrates. Acta Psychiatr Scand. 2005;111:94-105.
25. Henkel V, Seemuller F, Obermeier M, et al. Does early improvement of depressive syndromes triggered by antidepressant
compounds predict remission? Analysis of data of a naturalistic
study on a large sample of depressed inpatients. World J Biol
Psychiatry. In press.
26. Seemüller F, Riedel M, Obermeier M, et al. The controversial
link between antidepressants and suicidality risks in adults: do
data from the German Depression Katamnesis study support the
FDA decisions of suicide warnings for antidepressants? Eur Arch
Psychiatry Clin Neurosci. In press.
27. Seemuller F, Riedel M, Wickelmaier F, et al. Atypical symptoms in hospitalised patients with major depressive episode: frequency, clinical characteristics, and internal validity. J Affect
Disord. 2008;108:261-278.
28. Möller HJ, Langer S, Schmauss M. Escitalopram in clinical
practice: results of an open-label trial in outpatients with depression in a naturalistic setting in Germany. Pharmacopsychiatry.
2007;40:53-57.
29. Stassen HH, Angst J, Hell D, Scharfetter C, Szegedi A. Is there
a common resilience mechanism underlying antidepressant drug
response? Evidence from 2848 patients. J Clin Psychiatry. 2007;
68:1195-1205.
30. Quitkin FM, Rabkin JD, Markowitz JM, Stewart JW, McGrath
PJ, Harrison W. Use of pattern analysis to identify true drug response. A replication. Arch Gen Psychiatry. 1987;44:259-264.
31. Quitkin FM, McGrath PJ, Stewart JW, Taylor BP, Klein DF. Can
the effects of antidepressants be observed in the first two weeks
of treatment? Neuropsychopharmacology. 1996;15:390-394.
32. Nierenberg AA, Farabaugh AH, Alpert JE, et al. Timing of onset of antidepressant response with fluoxetine treatment. Am J
Psychiatry. 2000;157:1423-1428.
33. Stassen HH, Angst J, Delini-Stula A. Delayed onset of action
of antidepressant drugs? Survey of results of Zurich meta-analyses. Pharmacopsychiatry. 1996;29:87-96.
34. Szegedi A, Muller MJ, Anghelescu I, Klawe C, Kohnen R,
Benkert O. Early improvement under mirtazapine and paroxetine predicts later stable response and remission with high sensitivity in patients with major depression. J Clin Psychiatry.2003;
64:413-420.
35. Frodl T, Meisenzahl EM, Zetzsche T, et al. Hippocampal and
amygdala changes in patients with major depressive disorder and
healthy controls during a 1-year follow-up. J Clin Psychiatry.
2004;65:492-499.
IN
DEPRESSION: T
HE
ULTIMATE GOAL
36. Rojo JE, Gibert K, Cobo J, Rodriguez-Cano E, Vallejo J. Onset
of antidepressant action: a pharmacological question? Hum Psychopharmacol. 2005;20:425-433.
37. Klein DF. Listening to meta-analysis but hearing bias. Prevention & Treatment. 1998;1:Article ID 0006c. June 26,1998.
38. Möller HJ, Fischer G, von Zerssen D. Prediction of therapeutic response in acute treatment with antidepressants. Results of
an empirical study involving 159 endogenous depressive inpatients. Eur Arch Psychiatry Neurol Sci. 1987;236:349-357.
39. Seemüller F, Möller HJ, Obermeier M, et al. Prediction of response and remission in inpatients with depressive syndromes.
World J Biol Psychiatry. In press.
40. Sharan P, Saxena S. Treatment-resistant depression: clinical
significance, concept and management. Natl Med J India. 1998;
11:69-79.
41. Linden M. Therapeutic strategies for improving patient compliance (author’s translation from German). Nervenarzt.1979;50:
109-114.
42. Gilmer WS, Gollan JK, Wisniewski SR, et al. Does the duration of index episode affect the treatment outcome of major depressive disorder? A STAR*D report. J Clin Psychiatry. 2008;69:
1246-1256.
43. Uhr M, Tontsch A, Namendorf C, et al. Polymorphisms in the
drug transporter gene ABCB1 predict antidepressant treatment
response in depression. Neuron. 2008;57:203-209.
DÉLAI
DE RÉPONSE ET RÉMISSION AU COURS
DU TRAITEMENT ANTIDÉPRESSEUR
L
a dépression majeure est une maladie complexe et hétérogène dont l’étiopathogenèse est basée sur de nombreux facteurs qui peuvent agir à différents niveaux (psychologique, biologique, génétique et social). Il est
donc avéré qu’un traitement unique ou une approche thérapeutique unique
ne peut convenir à une maladie aussi diversifiée. Nous avons l’espoir dans un
futur proche de pouvoir traiter les patients de façon encore plus personnalisée
grâce aux marqueurs cérébraux morphologiques et génétiques. En cas de résistance au traitement, une pseudorésistance (en insistant sur l’observance)
doit être tout d’abord éliminée. La réponse au traitement peut être très précoce,
dès les 2 premières semaines du traitement, mais dans les études cliniques le
taux le plus élevé de réponses se situe autour des 4 premières semaines. Quand
exactement et dans quelle mesure la réponse apparaît, dépendent aussi de sa
définition exacte. La communauté scientifique a adopté un consensus sur deux
définitions : celle de l’amélioration précoce (20 % de réduction sur l’échelle de
HAM-D [Hamilton Rating Scale for Depression] au cours des 2 premières semaines) et celle de la réponse proprement dite ( 50 % de réduction du score
initial HAM-D). La première est surtout utile dans la surveillance du soulagement des symptômes grâce au traitement en question alors que la seconde est
plus utilisée pour la détection de modifications prolongées, particulièrement
en association avec un critère temporel. L’amélioration précoce et la réponse,
ainsi définies, sont fortement prédictives de rémission. Les études cliniques indiquent que si les taux de rémission sont habituellement plus faibles que ceux
de réponse, ils les dépassent néanmoins à partir de la 4e à la 6e semaine. La réponse au traitement apparaît donc comme un précurseur de la rémission, et
donc comme la phase initiale de l’évolution naturelle de la dépression sous
traitements antidépresseurs, qui dans la majorité des cas aboutira à la rémission, c’est à dire à la disparition complète de tous les symptômes.
Time course of response and remission during antidepressant treatment – Möller and others
MEDICOGRAPHIA, VOL 31, No. 2, 2009 125
IN
DEPRESSION: THE ULTIMATE GOAL
TRUE REMISSION
David S. BALDWIN, DM FRCPsych
Antonio T. V. LOPES, MB BS, MRCPsych
Clinical Neuroscience Division
School of Medicine, University of Southampton
Mood and Anxiety Disorders Service
Hampshire Partnership Trust
Southampton, UNITED KINGDOM
The influence of comorbid
anxiety disorders on outcome
in major depressive disorder
b y D . S . B a l d w i n a n d A . T. V. L o p e s ,
United Kingdom
nxiety symptoms are common in patients
experiencing major depressive episodes, being reported by approximately 60% of patients, and comorbid anxiety disorders are seen in
around 50% of patients with major depressive disorder.1 One of the early findings of the influential US
Sequenced Treatment Alternatives to Relieve Depression (STAR*D) study was that “anxious depression,” defined as a score of 7 or greater on the anxiety-somatization subscale of the Hamilton Rating
A
oexisting anxiety symptoms and comorbid anxiety disorders are common in patients with major depressive disorder. This review examines
three aspects of the relationship between major depressive disorder and
comorbid anxiety disorders: whether the comorbid condition is more severe
than “pure” major depression; whether the comorbid condition is associated
with worse clinical outcomes than are seen in major depressive disorder alone;
and whether the response to antidepressant treatment differs between depressed
patients with or without comorbid anxiety disorders. Although not all evidence
is consistent, in general terms, the presence of comorbid anxiety disorders in
patients with major depressive disorder is associated with greater severity of
symptoms and more pronounced impairment. The course of illness is less favorable in patients with the comorbid condition, and relatively fewer depressed
patients respond to antidepressant treatment and achieve remission of symptoms if affected by comorbid anxiety disorders. There is a need for randomized
placebo-controlled studies specifically in patients with comorbid major depressive disorder and anxiety disorders, in order to determine whether this patient
group differs from those with “pure” major depression in its responsiveness to
pharmacological or psychological interventions.
C
Medicographia. 2009;31:126-131.
(see French abstract on page 131)
Keywords: major depression; anxiety disorder; comorbid; outcome; response
www.medicographia.com
Address for correspondence: Professor David S. Baldwin, University Department of Mental Health,
Royal South Hants Hospital, Brinton’s Terrace, Southampton SO14 0YG, UK
(e-mail: [email protected])
126 MEDICOGRAPHIA, VOL 31, No. 2, 2009
Scale for Depression,2 was seen in 49% of the patients in primary care, and by 42% of those treated
in secondary care.3 Furthermore, a comorbid anxiety disorder was present in approximately half of
patients, regardless of the treatment setting.4 This
degree of comorbidity is also seen in nonclinical
samples: for example, the results of a systematic review of European community studies among people
aged 18-65 years found that approximately 30%40% of patients with a depressive disorder had a comorbid anxiety disorder, and vice versa.5 The recent
European Study of the Epidemiology of Mental
Disorders (ESEMeD) also found a high 12-month
prevalence of comorbid mood and anxiety disorders
(Figure 1).6
There have been few studies of the comorbidity
between anxiety and depressive disorders in later
life, although a Dutch community study of people
aged 55-85 years found that 47.5% of those with a
major depressive disorder also met criteria for at
least one anxiety disorder, and 26.1% of those with
anxiety disorders fulfilled the criteria for major depression.7 This observation is supported by the findings of studies involving clinical samples of older
depressed patients, in whom comorbid anxiety disorders have been found to vary in frequency from
between 3%-65%.8-10
SELECTED
ABBREVIATIONS AND ACRONYMS
DSM-IV TR Diagnostic and Statistical Manual of
Mental Disorders - Fourth Edition
Text Revision
ESEMeD
European Study of the Epidemiology
of Mental Disorders
SSRI
selective serotonin reuptake inhibitor
STAR*D
Sequenced Treatment Alternatives to
Relieve Depression (study)
The influence of comorbid anxiety disorders on outcome in major depressive disorder – Baldwin and Lopes
TRUE REMISSION
Greater severity of symptoms and
impairment in the comorbid condition
Early studies supported the widespread consensus
that patients with comorbid mood and anxiety disorders had more severe symptoms.11-14 Subsequent
studies have in general supported this view. For example, a comparison of 276 US primary care depressed patients with or without a lifetime comorbid
anxiety disorder demonstrated that the presence of
comorbid panic disorder was associated with greater
severity of depressive symptoms, more marked impairment in psychosocial functioning, and greater
risk of prematurely stopping treatment.15
Similar findings were seen in a more recent naturalistic study of the effects of coexisting anxiety
symptoms or comorbid anxiety disorders on symptom severity at baseline and treatment response, in
Italian outpatients with major depressive disorder.
It was found that the presence of anxiety symptoms
and disorders was associated with more frequent
suicidal thoughts, greater psychomotor retardation,
and greater severity of diurnal variation of symptoms, sexual dysfunction, somatic concerns, and
weight loss, when compared with patients with major depressive disorder alone.16
The greater symptom severity associated with the
presence of comorbid anxiety disorders is also seen
in other age groups: for example, a comparison of
the effectiveness of interpersonal psychotherapy
versus “treatment as usual” in depressed adolescents (aged 12-18 years) found that comorbidity was
associated with both a greater severity of depressive
symptoms at baseline, and with lower response rates
in both treatment groups.17
The greater symptom severity of the comorbid
condition is reflected in a more pronounced degree
of impairment of social and occupational functioning. For example, a recent large cross-sectional primary care study performed in Belgium and Luxembourg found that patients with comorbid major
depressive disorder and generalized anxiety disorder reported greater impairment of work, and social
and family life, than did patients with major depression or generalized anxiety disorder alone.18
Epidemiological studies demonstrate that a number of individuals have multiple comorbid diagnoses, and some of the greater impairment that is
associated with comorbid depressive and anxiety
disorders may reflect the presence of additional mental health problems. For example, the presence of
comorbidity for an anxiety disorder in patients with
major depressive disorder has also been associated
with greater risk of comorbidity for personality dis-
DEPRESSION: T
HE
ULTIMATE GOAL
orders: in the aforementioned comparison of US
primary care depressed patients, lifetime comorbidity for panic disorder was also associated with the
presence of avoidant personality disorder.15
The adverse effects of anxiety disorder comorbidity in patients with unipolar depressive disorder are
sometimes also seen in patients with the diagnosis
of bipolar disorder, although not all studies have
produced consistent findings. An investigation in
a consecutive sample of French bipolar inpatients
found that those with a lifetime comorbid anxiety
disorder (24% of the overall sample) did not differ
in terms of disorder severity (assessed by number of
hospitalizations, presence of psychosis, substance
misuse comorbidity, and suicide attempts), although
their response to anticonvulsant drugs was lower
than that of the group without comorbid anxiety
6.2
Men
Women
6
*P<0.05 by gender
5
Percentage (%)
This review examines three key aspects of the relationship between major depressive disorder and
comorbid anxiety disorders. First, whether the comorbid condition is more severe than “pure” major
depression; second, whether the comorbid condition is associated with worse clinical outcomes than
are seen in major depressive disorder alone; and
third, whether the response to antidepressant treatment differs between depressed patients with or
without comorbid anxiety disorders.
IN
4
3.1
3
2
2.8*
1.8*
2.4
1.4*
0.8*
1
0.2
0
Mood
Anxiety
Alcohol
0.1* 0.0
Mood
+ Anxiety
Pure disorders
0.1 0.0
0.1* 0.1
Mood
Anxiety
Mood
+ Alcohol + Alcohol + Anxiety
+ Alcohol
Comorbid disorders
Figure 1. Prevalence of pure and comorbid 12-month mood, anxiety, and alcohol use
disorders in the general population. Findings from the European Study of the Epidemiology of Mental Disorders.
After reference 6: Alonso J, Angermeyer MC, Bernert S, et al. Prevalence of mental disorders in Europe:
results from the European Study of the Epidemiology of Mental Disorders (ESEMeD). Acta Psychiatrica
Scand. 2004;109(suppl 420):21-27. Copyright © 2004, John Wiley & Sons, Inc.
disorders.19 By contrast, in a longitudinal study of
the effects of comorbid anxiety disorders on bipolar
disorder patients treated with psychotropic drugs,
either alone or in combination with family intervention, comorbidity was associated with greater
symptom severity at baseline (even after controlling
for depressive symptom severity) and with poorer
overall treatment response over 28 months, regardless of the treatment modality.20 The link between
bipolar disorder and anxiety disorders is also emphasized by the findings of an investigation of the
factors associated with non-response to antidepressant treatment in patients with apparent unipolar
depression, which found that the presence of comorbid anxiety disorders was associated with a
greater “risk” of unrecognized bipolar disorder.21
Poorer outcome in longitudinal studies
in patients with comorbid conditions
The presence of comorbid anxiety disorders with
major depression is usually found to be associated
with a less favorable long-term outcome. For exam-
The influence of comorbid anxiety disorders on outcome in major depressive disorder – Baldwin and Lopes
MEDICOGRAPHIA, VOL 31, No. 2, 2009 127
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
ple, an early systematic review of the clinical outcome of anxiety and depressive disorders found that
patients with comorbid anxiety and depression had
generally worse outcomes than patients with either
an anxiety disorder alone, or a depressive disorder
alone.22
The findings of the United States National Comorbidity Survey indicate that participants were
significantly more likely to continue presenting
with symptoms of a major depressive if they also
fulfilled criteria for comorbid generalized anxiety
disorder. Similarly, patients with generalized anxiety disorder and comorbid major depression were
more likely to experience continued anxiety symptoms than those without depressive symptoms
(Figure 2).23
2.0
*P<0.05
Odds ratio
1.8
1.2
0.9*
1.0
0.8*
0.0
With
MDE
Without
MDE
GAD
With
GAD
Without
GAD
MDE
Figure 2. Persistence of comorbid major depressive
disorder and generalized anxiety disorder (GAD).
MDE, major depressive episode.
After reference 23: Kessler RC, Gruber M, Hettema JM, Hwang I,
Sampson N, Yonkers KA. Co-morbid major depression and generalized anxiety disorders in the National Comorbidity Survey follow-up.
Psychological Med. 2008;38:365-374. Copyright © 2007, Cambridge
University Press.
In general terms, this adverse effect of comorbidity is manifest through longer persistence of symptoms and with a greater risk of continuing social
and occupational impairment. For example, an evaluation of the effects of comorbid post-traumatic
stress disorder in female patients with major depressive disorder treated with either antidepressant
drugs, cognitive behavior therapy, or through community health referral, found that comorbidity was
associated not only with greater severity of depressive and anxiety symptoms at baseline, but also
with greater impairment at 1-year follow-up.24 This
adverse effect of this pattern of comorbidity on the
clinical outcomes supports the earlier findings of
a delayed response to treatment of depression in
both the acute phase25 and the continuation phase26
of treatment, in depressed patients with comorbid
post-traumatic disorder.
The adverse effects of coexisting anxiety symptoms or comorbid anxiety disorder are also seen in
older depressed patients. In a study of the effectiveness of antidepressant treatment and interpersonal psychotherapy in patients aged 70 years or older,
the presence of greater anxiety symptom severity
128 MEDICOGRAPHIA, VOL 31, No. 2, 2009
was associated with a longer duration of symptoms
and with higher recurrence rates.27 Similar findings
had been seen in an earlier investigation of the
course of illness in elderly patients with comorbid
major depression and generalized anxiety disorder.28
The adverse effects of comorbidity are also manifest through a greater risk of recurrence of symptoms and possibly through an increased risk of suicide. By way of illustration, long-term follow-up
(up to 5 years) of a Finnish nationally representative sample of outpatients with DSM-IV (Diagnostic
and Statistical Manual of Mental Disorders - Fourth
Edition)–defined major depressive disorder found
that the severity of symptoms and the presence of
comorbidity, especially for social phobia, predicted
both a higher probability of recurrence and a greater
number of recurrences.29
While the increased risk of suicide in depression
has been known for many years, it remained uncertain as to whether anxiety disorders were also associated with an elevated risk of suicide. However, in
recent years, epidemiological studies and a systematic review have demonstrated that anxiety disorders are associated with suicidal thoughts and with
attempted, if not completed, suicide.30-32 Moreover,
current data indicate that comorbid anxiety disorders amplify the risk of suicide attempts in people
with mood disorders.30 As the two greatest risk factors for completed suicide are suicidal thoughts and
a recent suicide attempt, it therefore seems reasonable to assume that anxiety disorders carry an increased risk for completed suicide.
Reduced response rates and less
remission of symptoms during treatment
The presence of prominent anxiety symptoms in depressed patients is generally thought to be associated with a lower overall response rate to treatment.
For example, an early comparison of the effectiveness of interpersonal psychotherapy or treatment
with the tricyclic antidepressant nortriptyline in
157 depressed patients in primary care found that
lifetime comorbidity for an anxiety disorder was associated with higher treatment drop-out rates, a delayed recovery in patients with comorbid generalized anxiety disorder, and particularly poor response
rates in patients with lifetime panic disorder.33
Subsequently, a comparison of small groups of
patients with major depressive disorder that did or
did not respond to antidepressant treatment found
that comorbid anxiety disorders (and reports of
childhood emotional abuse) were significantly more
frequent among the nonresponders.34 A much larger randomized trial of stepped collaborative care of
228 depressed patients in a US health maintenance
organization found that the presence of comorbid
panic disorder was associated with both greater
symptom severity at baseline, and with a significantly lower treatment response.35
Furthermore, in a recent investigation of the determinants of non-response in patients with treatment-resistant depression, a comorbid anxiety disorder was the clinical factor most strongly associated with non-response.36
The influence of comorbid anxiety disorders on outcome in major depressive disorder – Baldwin and Lopes
TRUE REMISSION
The adverse effects of comorbidity on the response
to treatment are seen in a variety of age groups. For
example, an evaluation of the effectiveness of cognitive behavioral therapy, systemic behavioral therapy, and nondirective supportive therapy in 101 adolescents (aged 13 to 18 years) fulfilling DSM-III-R
criteria for major depression found that the presence of anxiety disorder comorbidity was predictive
of symptom persistence.37 At the other end of the
age distribution, a longitudinal study in older adults
undergoing case management, cognitive behavior
therapy, or the combination for treatment of depression found that comorbid anxiety disorders were associated with greater symptom severity at the end
of treatment and at follow-up at 6 and 12 months.38
However, not all the evidence for the effect of coexisting anxiety symptoms or comorbid disorders
on treatment response is consistent. For example, a
comparison of small groups of Australian inpatients
with DSM-III-R–defined major depression found
that the presence of comorbid anxiety disorders
did not affect either treatment choice, or the effectiveness of treatment interventions.39 In addition, a
greater severity of coexisting anxiety symptoms at
baseline among patients with either chronic major
depression or “double depression” (that is, dysthymia plus supervening acute major depression) did
not affect overall response rates with either the tricyclic imipramine or the selective serotonin reuptake inhibitor (SSRI) sertraline.40 Furthermore, in
both a comparison of the effectiveness of the tricyclic nortriptyline and the SSRI paroxetine in 116
depressed patients aged 60 years or older, and a second comparison of the effectiveness of paroxetine or
interpersonal psychotherapy in 125 patients aged 69
years or older, no difference was found in the proportion responding to treatment or in the time to response, between patients with or without anxiety.41
Although outside the scope of this review, it is
worth noting that the adverse effects of comorbidity are also seen among patients with primary anxiety disorders. For example, a recent evaluation of
clinical outcomes at 1 year following cognitive behavioral therapy in outpatients with panic disorder,
with or without agoraphobia, found that the presence of comorbid mood disorders was associated
with lower response rates and a reduction in the
proportion entering symptomatic remission.42
Comorbidity of major depressive disorder with
anxiety disorders has also been associated with a reduced likelihood of achieving symptomatic remission with antidepressant treatments. The findings of
STAR*D, sequential treatment of 2876 depressed
patients, found that the presence of a comorbid anxiety disorder at baseline was associated with significantly lower rates of achieving symptomatic remission during the initial intervention (with the SSRI,
citalopram).43 However, as with studies of overall
treatment response, not all evidence is consistent:
for example, an evaluation of the effects of comorbid anxiety disorders on response to treatment with
the SSRI fluoxetine in 329 patients with DSM-IV–
defined major depressive disorder, found no major
adverse effects of comorbidity on the likelihood of
achieving symptomatic remission.44
IN
DEPRESSION: T
HE
ULTIMATE GOAL
Lower response rates and reduced likelihood of
achieving symptom remission are factors that may
lead to a greater perceived need to utilize antidepressant drugs, and to the concomitant use of two
or more antidepressant or other psychotropic drugs,
in an attempt to improve outcomes. Data from the
recent Canadian Community Health Survey on
Mental Health and Well-Being show that the comorbidity of major depressive disorder with anxiety
disorders is associated with somewhat higher rates
of use of antidepressant drugs than that seen for
major depressive disorder alone.45
The need for studies
in patients with comorbid mood
and anxiety disorders
The presence of comorbid depressive disorders is
usually regarded as an exclusion criterion in randomized placebo-controlled trials of antidepressants
in patients with primary anxiety disorders, where
proof of efficacy requires the demonstration that
anxiety symptoms do not resolve indirectly, mediated by an effect on depression. In addition, depressed
patients with comorbid anxiety disorders are often
excluded from participating in studies with new antidepressant treatments, perhaps because the presence of significant anxiety symptoms may reduce
response rates, and thereby impede the chance to
distinguish antidepressant from placebo effects.
However, comorbidity is the rule in clinical practice, and it is helpful to know whether a single medication can diminish the severity of both anxiety
and depressive symptoms in patients with comorbid
conditions. However, there are relatively few studies that have specifically focused on investigation of
the treatment response in comorbid patients.
An investigation of the effectiveness of open treatment with the SSRI fluoxetine (20 mg/day) in 123
US outpatients with major depressive disorder and
at least one comorbid anxiety disorder, both defined
according to DSM-III-R criteria, found that it reduced depressive symptom severity between baseline and study end point: however, patients with
depression and comorbid obsessive-compulsive disorder were significantly less likely to respond to
treatment than patients with other comorbid anxiety disorders.46 A subsequent open but controlled
comparison of paroxetine (20-40 mg/day) with moclobemide (a reversible inhibitor of monoamine oxidase A) at a dosage of 300-600 mg/day in 123 Italian
outpatients with DSM-III-R–defined major depressive disorder or dysthymia and comorbid anxiety
disorder found no significant difference in overall
response between the two antidepressants: however
paroxetine was superior to moclobemide in the subgroup of 32 patients with comorbid panic disorder.47
The open design and lack of placebo control in
these investigations together prevent definitive conclusions being drawn about the potential efficacy
of certain antidepressant treatments in comorbid
patients. A randomized double-blind comparatorcontrolled trial of sertraline (50-100 mg/day) and
imipramine (100-200 mg/day) in patients with comorbid major depressive disorder and panic disor-
The influence of comorbid anxiety disorders on outcome in major depressive disorder – Baldwin and Lopes
MEDICOGRAPHIA, VOL 31, No. 2, 2009 129
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
Anxiety symptoms are integral to major depressive
episodes, and many patients with major depressive
disorder will have prominent anxiety symptoms:
furthermore, a significant proportion of depressed
patients will show either lifetime or concurrent comorbidity for anxiety disorders. In general terms,
the presence of comorbid anxiety disorders is associated with a greater severity of symptoms and
more pronounced symptom-related disability and
impairment, with a less favorable outcome, greater
risk of symptom persistence, recurrence, and possibly suicide, and a less satisfactory response to antidepressant treatment. It is also associated with
lower rates of recovery and a reduced likelihood of
achieving symptomatic remission. There is a clear
need for further randomized controlled trials in patients with comorbid mood and anxiety disorders,
as this group comprises a probable majority of depressed individuals seen within routine clinical
practice settings. REFERENCES
1. Fava M, Uelbacker L, Alpert JE, Nierenberg AA, Pava JA, Rosenbaum JF. Major depressive subtypes and treatment response.
Biol Psychiatry. 1997;42:568-576.
2. Hamilton M. Development of a rating scale for a primary depressive illness. Br J Soc Clin Psychol. 1967;6:278-296.
3. Fava M, Rush AJ, Alpert JE, et al. What clinical and symptom
features and comorbid disorders characterize outpatients with
anxious major depressive disorder: a replication and extension.
Can J Psychiatry. 2006;51:823-835.
4. Gaynes BN, Rush AJ, Trivedi MH, et al. Major depression symptoms in primary care and psychiatric settings: a cross-sectional
analysis. Ann Fam Med. 2007;5:126-134.
5. Wittchen HU, Jacobi F. Size and burden of mental disorders in
Europe a critical review an appraisal of 27 studies. Eur Neuropsychopharmacol. 2005;15:357-376.
6. Alonso J, Angermeyer MC, Bernert S, et al. Prevalence of mental disorders in Europe: results from the European Study of the
Epidemiology of Mental Disorders (ESEMeD). Acta Psychiatrica
Scand. 2004;109(suppl 420):21-27.
7. Beekman AT, de Beurs E, van Balkom AJ, Deeg DJ, van Dyck R,
van Tilburg W. Anxiety and depression in later life: co-occurrence
and communality of risk factors. Am J Psychiatry. 2000;157:
89-95.
8. Parmalee P, Katz I, Lawton MD. Anxiety and its association
with depression among institutionalized elderly. Am J Geriatr
Psychiatry. 1993;1:65-78.
9. Mulsant BH, Reynolds CF, Shear MK, et al. Comorbid anxiety
disorders in late-life depression. Anxiety. 1996;2:242-247.
10. Lenze EJ, Mulsant BH, Shear MK, et al. Comorbid anxiety disorders in depressed elderly patients. Am J Psychiatry. 2000;157:
722-728.
11. Andrade L, Eaton WW, Chilcoat H. Lifetime comorbidity of
panic attacks and major depression in a population-based study.
Symptom profiles. Br J Psychiatry. 1994;165:363-369.
12. Grunhaus L, Pande AC, Brown MB, Greden JF. Clinical characteristics of patients with concurrent major depressive disorder
and panic disorder. Am J Psychiatry. 1994;151:541-546.
13. Kessler RC, McGonagle KA, Zhao S, et al. Lifetime and 12month prevalence of DSM-III-R psychiatric disorders in the United States. Results from the National Comorbidity Survey. Arch
Gen Psychiatry. 1994;51:8-19.
14. Brown TA, Antony MM, Barlow DH. Diagnostic comorbidity
in panic disorder: effect on treatment outcome and course of comorbid diagnoses following treatment. J Consult Clin Psychol.
1995;63:408-418.
15. Brown C, Schulberg HC, Shear KM. Phenomenology and
severity of major depression and comorbid lifetime anxiety disorders in primary medical care practice. Anxiety. 1996;2:210-218.
16. Altamura CA, Montresor C, Salvadori D, Mundo E. Does comorbid subthreshold anxiety affect clinical presentation and
treatment response in depression? A preliminary 12-month naturalistic study. Int J Neuropsychopharmacol. 2004;7:481-487.
17. Young JF, Mufson L, Davies M. Impact of comorbid anxiety
in an effectiveness study of interpersonal psychotherapy for depressed adolescents. J Acad Child Adolesc Psychiatry. 2006;45:
904-912.
18. Ansseau M, Fischler B, Dierick M. Albert A, Leyman S, Mignon
A. Socioeconomic correlates of generalized anxiety disorder and
major depression in primary care: the GADIS II Study (Generalized Anxiety and Depression Impact Survey II). Depress Anxiety.
2008;25:506-513.
19. Henry C, Van den Bulke D, Bellivier F, Etan B, Rouillon F,
Leboyer M. Anxiety disorders in 318 bipolar patients: prevalence
and impact on illness severity and response to mood stabilizer.
J Clin Psychiatry. 2003;64:331-335.
20. Gaudiano BA, Miller IW. Anxiety disorder comorbidity in bipolar I disorder: relationship to depression severity and treatment
outcome. Depress Anxiety. 2005;21:71-77.
21. Calabrese JR, Muzina DJ, Kemp DE, et al. Predictors of bipolar disorder among patients currently treated for major depression. Med Gen Med. 2006;8:38.
22. Emmanuel J, Simmonds S, Tyrer P. Systematic review of the
outcome of anxiety and depressive disorders. Br J Psychiatry.
1998;34(suppl):S35-S41.
23. Kessler RC, Gruber M, Hettema JM, Hwang I, Sampson N,
Yonkers KA. Co-morbid major depression and generalized anxiety disorders in the National Comorbidity Survey follow-up.
Psychological Med. 2008;38:365-374.
24. Green BL, Krupnick JL, Chung J, et al. Impact of PTSD comorbidity on one-year outcomes in a depression trial. J Clin Psychology. 2006;62:815-835.
25. Hegel MT, Unutzer J, Tang L, et al. Impact of comorbid panic and posttraumatic stress disorder on outcomes of collaborative care for late-life depression in primary care. Am J Geriatr Psychiatry. 2005;13:48-58.
26. Hollon SD, DeRubeis RJ, Shelton RC, et al. Prevention of relapse following cognitive therapy vs medications in moderate to
severe depression. Arch Gen Psychiatry. 2005;62:417-422.
27. Andreescu C, Lenze EJ, Dew MA, et al. Effect of comorbid anxiety on treatment response and relapse risk in late-life depression:
controlled study. Br J Psychiatry. 2007;190:344-349.
28. Schoevers RA, Deeg DJ, van Tilburg W, Beekman AT. Depression and generalized anxiety disorder: co-occurrence and longitudinal patterns in elderly patients. Am J Geriatr Psychiatry.
2005;13:31-39.
29. Holma KM, Holma IA, Melartin TK, Rytsala HJ, Isometsa ET.
Long-term outcome of major depressive disorder in psychiatric
patients is variable. J Clin Psychiatry. 2008;69:196-205.
30. Sareen J, Cox BJ, Agifi TO, et al. Anxiety disorders and risk for
suicidal ideation and suicide attempts: a population based longitudinal study of adults. Arch Gen Psychiatry. 2005;62:1249-1257.
31. Bolton JM, Belik SL, Enns MW, Cox BJ, Sareen J. Exploring
the correlates of suicide attempts among individuals with major
depressive disorder: findings from the national epidemiologic survey on alcohol and related conditions. J Clin Psychiatry. 2008;69:
1139-1149.
32. Hawgood J De LD. Anxiety disorders and suicidal behaviour:
an update. Curr Opin Psychiatry. 2008;21:51-64.
33. Brown C, Schulberg HC, Madonia MJ, Shear MK, Houck PR.
Treatment outcomes for primary care patients with major depression and lifetime anxiety disorders. Am J Psychiatry. 1996;153:
1293-1300.
34. Kaplan MJ, Klinetob NA. Childhood emotional trauma and
chronic posttraumatic stress disorder in adult outpatients with
treatment-resistant depression. J Nerv Ment Dis. 2000;188:596601.
35. Walker EA, Katon WJ, Russo J, et al. Predictors of outcome in
a primary care depression trial. J Gen Intern Med. 2000;15:859867.
36. Souery D, Oswald P, Massat I, et al; Group for the Study of
Resistant Depression. Clinical factors associated with treatment
resistance in major depressive disorder: results from a European
der found no differences between treatments in the
reduction of depressive and panic symptom severity, treatment outcome being concordant for both
diagnoses in approximately 70% of patients.48 Again,
the absence of a placebo control hinders definitive
conclusions, and there is a persistent need for large
multicenter studies in patients with comorbid mood
and anxiety disorders, that employ a placebo-controlled design.
Conclusions
130 MEDICOGRAPHIA, VOL 31, No. 2, 2009
The influence of comorbid anxiety disorders on outcome in major depressive disorder – Baldwin and Lopes
TRUE REMISSION
multicenter study. J Clin Psychiatry. 2007;68:1062-1070.
37. Brent DA, Kolko DJ, Birmaher B, et al. Predictors of treatment efficacy in a clinical trial of three psychosocial treatments
for adolescent depression. J Acad Child Adolesc Psychiatry. 1998;
37:906-914.
38. Gum AM, Arean PA, Bostrom A. Low-income depressed older adults with psychiatric comorbidity: secondary analyses of response to psychotherapy and case management. Int J Geriatr Psychiatry. 2007;22:124-130.
39. Rodney J, Prior N, Cooper B, et al. The comorbidity of anxiety and depression. Aust NZ J Psychiatry. 1997;31:700-703.
40. Hirschfeld RM, Russell JM, Delgado PL, et al. Predictors of response to acute treatment of chronic and double depression with
sertraline or imipramine. J Clin Psychiatry. 1998;59:669-675.
41. Lenze EJ, Mulsant BH, Dew MA, et al. Good treatment outcomes in late-life depression with comorbid anxiety. J Affect Disord. 2003;77:247-254.
42. Van Balkom AJ, van Boeijen CA, Boeke AJ, van Oppen P,
Kempe PT, van Dyck R. Comorbid depression, but not comorbid
anxiety disorders, predicts poor response in anxiety disorders. Depress Anx. 2008;25:408-415.
IN
DEPRESSION: T
HE
ULTIMATE GOAL
43. Trivedi MH, Rush AJ, Wisniewski SR, et al; STAR*D Study
Team. Evaluation of outcomes with citalopram for depression
using measurement-based care in STAR*D: implications for clinical practice. Am J Psychiatry. 2006;163:28-40.
44. Cassano P, Soares CN, Cohen LS, Lyster AK, Fava M. Sex- and
age-related differences in major depressive disorder treated with
fluoxetine. Arch Womens Ment Health. 2004;7:167-171.
45. Schaffer A, Cairney J, Veldhuizen S, Cheung A, Levitt A. Comparison of antidepressant use between subjects with bipolar disorder and major depressive disorder with or without comorbid
anxiety. J Clin Psychiatry. 2007;68:1785-1792.
46. Lepola U, Arato M, Zhu Y, Austin C. Sertraline versus imipramine treatment of comorbid panic disorder and major depressive disorder. J Clin Psychiatry. 2003;64:654-662.
47. Pini S, Amador XF, Dell’Osso L, et al. Treatment of depression
with comorbid anxiety disorders: differential efficacy of paroxetine versus moclobemide. Int Clin Psychopharmacol. 2003;18:
15-21.
48. Sonawalla SB, Farabaugh A, Johnson MW, et al. Fluoxetine
treatment of depressed patients with comorbid anxiety disorders.
J Psychopharmacol. 2002;16:215-219.
IMPACT
DES TROUBLES ANXIEUX COMORBIDES
SUR LA RÉMISSION DE LA DÉPRESSION
L
a coexistence de symptômes anxieux et de troubles anxieux comorbides
est fréquente chez les patients atteints de troubles dépressifs majeurs
(TDM). Cet article étudie trois aspects de la relation entre les TDM et les
troubles anxieux comorbides : lorsque l’état comorbide est plus sévère que la
dépression majeure « pure » ; lorsque l’évolution clinique de l’état comorbide
est plus sévère que celle des TDM isolés ; et lorsque la réponse au traitement
antidépresseur diffère entre les patients avec ou sans troubles anxieux comorbides associés. En général, même si les données ne sont pas toutes concordantes,
la présence de troubles anxieux comorbides chez les patients atteints de TDM
est associée à une plus grande sévérité des symptômes et à une atteinte plus
prononcée. L’évolution de la maladie est moins favorable chez les patients atteints de comorbidité et un plus petit nombre d’entre eux répondent au traitement antidépresseur et obtiennent une rémission de leurs symptômes. Des
études randomisées contrôlées contre placebo sont nécessaires chez les patients
souffrant de TDM et de troubles anxieux comorbides afin de déterminer si ces
derniers diffèrent de ceux souffrant de dépression majeure « pure » dans la réponse aux traitements pharmacologiques et psychologiques.
The influence of comorbid anxiety disorders on outcome in major depressive disorder – Baldwin and Lopes
MEDICOGRAPHIA, VOL 31, No. 2, 2009 131
TRUE REMISSION
IN
DEPRESSION: THE ULTIMATE GOAL
Palmiero MONTELEONE, MD
Mario MAJ, MD, PhD
Department of Psychiatry
University of Naples SUN
Naples, ITALY
Circadian rhythm
disturbances in depression:
implications for treatment and
quality of remission
b y P. M o n t e l e o n e a n d M . M a j , I t a l y
ver the course of evolution, organisms have
developed cellular clock mechanisms sensitive to light, and have adapted by organizing
their activities in 24-hour cycles determined by sunrise and sunset. These cycles do not simply reflect
an organism’s passive response to environmental
changes, such as the light-dark cycle, but rather
O
ood disorders, especially unipolar depression and seasonal affective
disorder, have been linked to endogenous circadian rhythm abnormalities. Evidence is emerging that disruption of the normal circadian
rhythmicity occurs at least in a subgroup of depressed patients, although whether
there is a causal link between endogenous circadian rhythm disruption and
depression has not been firmly demonstrated. Nonetheless, improvements in
some forms of depression in response to strategies that manipulate circadian
rhythms support the idea that circadian abnormalities observed in depressed
patients may constitute a core component of the pathophysiology of depression
and are worthy of therapeutic consideration. Chronotherapeutic interventions,
which include both nonpharmacological strategies, such as sleep deprivation,
light therapy, and interpersonal and social rhythm therapy, and pharmacological treatments based on the use of drugs specifically endowed with chronobiotic properties, such as agomelatine, have been shown to have antidepressant
effects. Therefore, normalization of circadian rhythms seems to represent a possible new direction for the development of either pharmacological or nonpharmacological innovative therapeutic strategies, which could have an important
future role as alternatives or adjuvants to currently available antidepressant
treatments, in order to achieve a better quality of remission and a persistent
amelioration of patients’ social functioning and quality of life.
M
Medicographia. 2009;31:132-139.
(see French abstract on page 139)
Keywords: circadian rhythm; depression; remission; chronotherapy;
antidepressant
www.medicographia.com
Address for correspondence: Professor Palmiero Monteleone, Department of Psychiatry,
University of Naples SUN, Largo Madonna delle Grazie, 80128 Naples, Italy
(e-mail: [email protected])
132 MEDICOGRAPHIA, VOL 31, No. 2, 2009
represent pre-adapted endogenous rhythms, which
arise from a timekeeping system within the organism and that persist in the absence of any environmental stimuli. The endogenous timekeeping
system or biological clock allows the organism to
anticipate and prepare for the changes in the environment that are associated with day and night in
order to function optimally.
In humans and other mammals, circadian rhythms
are generated by an internal clock or pacemaker,
which is located in the suprachiasmatic nucleus
(SCN) of the hypothalamus.1 Individual neurons
from the SCN, when dissociated and held in vitro,
retain a robust circadian rhythm in electrical firing,
and this can continuously be recorded for several
weeks, showing a slight deviation from 24 hours
(usually longer).2 Thus, the intrinsic rhythmicity
of the endogenous clock requires daily synchronization to the 24-hour day by regularly occurring environmental signals or “zeitgebers.” Light, the major zeitgeber for the SCN, reaches the SCN neurons
SELECTED
ABBREVIATIONS AND ACRONYMS
DSM-IV Diagnostic and Statistical Manual of
Mental Disorders–Fourth Edition
HPA
hypothalamo-pituitary-adrenal (axis)
IPSRT interpersonal and social rhythm therapy
PVN
paraventricular nucleus
REM
rapid eye movement
SAD
seasonal affective disorder
SCN
suprachiasmatic nucleus
SNP
single nucleotide polymorphism
STAR*D Sequenced Treatment Alternatives to
Relieve Depression (study)
SWS
slow-wave sleep
TSH
thyroid stimulating hormone
Circadian rhythm disturbances in depression: remission quality/treatment implications – Monteleone and Maj
TRUE REMISSION
directly via the retinohypothalamic tract, a non–
image-forming pathway, and indirectly via the intergeniculate leaflet of the lateral geniculate complex. The activity of SCN neurons is also modulated
by serotonin (5-HT) released by nervous fibers ascending from the raphe nuclei3 and by melatonin
secreted in the pineal gland.4
The major output of the SCN is to the paraventricular nucleus (PVN) of the hypothalamus, and
via a multisynaptic pathway, to the pineal gland,
where melatonin is synthesized according to the
length of the photoperiod. In this way, melatonin is
secreted at night and suppressed by light during
the day. Melatonin is a biochemical transducer of
photoperiodic information to all cells in the body
(including SCN neurons), signaling the seasonal
variations of the day/night cycle length.5 The PVN
is also the site of autonomic neurons, which communicate the time-of-day signal to different body
organs, and corticotrophin-releasing factor–secreting neurons, which are part of the hypothalamopituitary-adrenal (HPA) axis, endowed with a diurnal rhythmicity. Therefore, the SCN time signal is
translated into hormonal and autonomic signals
for peripheral organs mainly within the PVN.
Beside light there are also other nonphotic zeitgebers (among them exercise, food availability, temperature, jobs, and social demands) and these also
act directly or indirectly on the SCN to synchronize
its rhythmic activity. Furthermore, it is now widely demonstrated that multiple endogenous clocks
are distributed in every organ and perhaps in every
cell of the organism, and each of them has its own
zeitgebers.6
Nearly all physiological and behavioral functions
in humans are rhythmic, and examples to be mentioned here are the secretion patterns of hormones
(prolactin, corticotrophin, cortisol, growth hormone, melatonin), the sleep-wake cycle, core body
temperature, thyroid function, urine output, and
bronchial smooth muscle reactivity. These rhythms
enable the organism to synchronize endogenous
processes of the internal milieu and to anticipate
the periodic fluctuations in its external environment, with the aim of optimally dealing with them.
Circadian rhythm disturbances
and depression
IN
DEPRESSION: T
HE
ULTIMATE GOAL
of mood episodes, and on the other hand, approximately 20% of depressive patients show marked
diurnal mood swings. In a recent post-hoc analysis
of the Sequenced Treatment Alternatives to Relieve
Depression (STAR*D) study, it was reported that
21.6% of the depressed patients enrolled in the study
experienced diurnal mood variation, and that compared with patients without diurnal mood variation,
they exhibited more severe depression and were
more likely to meet the criteria for the melancholic
subtype of depression.11 Of note, a pattern of worse
mood in the morning is incorporated into the for-
Some patients with mood disorders suffer from regular cycles of altered mood
recurrence
Approximately 20% of depressed patients have marked diurnal variation in
mood,11 and a pattern of worse mood in the morning is incorporated into the
formal DSM-IV criteria for the melancholic subtype of major depression
Diurnal mood variations in major depressive patients are paralleled by diur-
nal variations in regional brain glucose metabolism12
A certain number of depressed patients exhibit elevated nocturnal body tem-
perature, with a phase advance in the overall 24-hour pattern13-16
The mean plasma concentrations of thyroid stimulating hormone during
sleep, its nocturnal peak, and the amplitude of its circadian rhythm have been
reported to be lower in depressed subjects, with a phase advance of the nocturnal peak17
Individuals with depression, especially those with the melancholic subtype,
show an overall increased cortisol secretion with a phase advance of the cortisol circadian rhythm18
Some patients with major depression show a lower blood concentration of
melatonin and a phase advance, or trend toward a phase advance, of the melatonin circadian rhythm19,20
It has been demonstrated that in bipolar patients and their offspring, the sen-
sitivity of melatonin to the suppressant effect of light is modified21,22
Most individuals with depression make subjective complaints about sleep-
wake cycle alterations; they also show sleep architecture abnormalities23-26
A phase advance of the circadian motor activity rhythm has been detected in
bipolar disorder patients during both the depressive and manic phases, as
well as during euthymia27-29
Patients with seasonal affective disorder regularly suffer major depressive
episodes that occur in the fall/winter
Polymorphic variants of clock genes have been found to be associated with
mood disorders, especially bipolar disorder30-38
Sleep deprivation and light therapy have proven antidepressant effects39-45
Interpersonal and social rhythm therapy has shown preliminary effects in the
prevention of mood episodes in bipolar patients46,47
Given that most human functions demonstrate circadian rhythmicity, it is intuitive that alterations in
the endogenous machinery regulating biorhythms
may lead to both physical and mental disorders.7-9
In particular, disruptions of endogenous biological
rhythms have been strongly associated with mood
disorders, especially unipolar depression, for which
alterations of circadian rhythms were first described
more than 20 years ago.10 There are several lines of
evidence supporting the occurrence of a dysregulation of the endogenous clock system in subgroups
of affective patients (Table I).11-53
Diurnal mood variation
First of all, on the one hand, some affective disorder patients suffer from regular cycles of recurrence
Data with a novel antidepressant drug, agomelatine, point to a restoration of
disrupted circadian rhythms in both animals and humans48-53
Table I. Evidence in support of a dysregulation of the endogenous clock system in
some patients with mood disorders.
mal DSM-IV (Diagnostic and Statistical Manual of
Mental Disorders–Fourth Edition) criteria for the
melancholic subtype of major depression, although
in the post-hoc STAR*D study analysis, diurnal
mood variation was more meaningfully related to
symptoms of melancholia if the definition was expanded to include also afternoon and evening worsening of mood.11 Also, patients with other forms of
depression actually appear to report a pattern of diurnal mood variation.54 In a recent investigation
Circadian rhythm disturbances in depression: remission quality/treatment implications – Monteleone and Maj
MEDICOGRAPHIA, VOL 31, No. 2, 2009 133
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
using a two-factor model of mood categorized according to positive affect and negative affect, diurnal variations in these two mood dimensions were
compared between a group of patients with major
depression and a group of healthy controls.55 Results indicated that depressed patients showed lower overall levels of positive affect, which increased
over the course of the day as in healthy controls but
with a backward-shifted acrophase, and higher overall negative affect levels, with maximum values occurring in the late morning and then decreasing
over the rest of the day. Furthermore, Germain et
al12 provided preliminary evidence that diurnal mood
variations in major depressive disorder patients were
paralleled by diurnal variations in regional brain
glucose metabolism.
Core body temperature
For core body temperature, a robust circadian
rhythm has been well established, with the highest values occurring in the evening and the nadir
occurring during the last third of the night. An elevated nocturnal body temperature is the most consistently observed circadian abnormality in depression,13,56 and this aberration generally normalizes
with clinical improvement.57 Although not confirmed by all studies, a phase-advance in the overall 24-hour pattern of body temperature has been
also reported in many depressed patients.13-16 Moreover, in subjects experiencing a depressive episode,
nighttime changes in body temperature have been
found to be inversely correlated with nighttime
changes in plasma levels of thyroid stimulating hormone (TSH).13 The mean plasma concentration of
TSH during sleep, its nocturnal peak concentration, and the amplitude of its circadian rhythm have
been reported to be lower in depressed subjects
compared with both normal controls and remitted
patients.17 Finally, the time of the nocturnal TSH
peak has been found to be advanced during a depressive episode.17
Cortisol secretion
In healthy subjects, maximal secretion of cortisol
occurs in the morning; thereafter, there is a progressive decline over the day until the nadir is
reached in the evening, immediately after falling
asleep. Dysregulation of the HPA axis is extremely
frequent in depressed patients. A meta-analysis on
cortisol in depression revealed an overall increase in
cortisol secretion, with the largest effect at the nadir
of the circadian rhythm and an earlier onset of the
first cortisol secretory episode, consistent with a
phase-advance of the cortisol circadian rhythm in
depression.18
Melatonin secretion and the sleep-wake cycle
Several studies have also reported on alterations in
the melatonin secretory pattern in depression: the
most consistent finding has been a lower blood concentration of melatonin and a phase advance, or a
trend toward a phase advance, of the melatonin circadian rhythm in individuals suffering from major depression19,20; this was, however, not confirmed
in all studies.58 Moreover, a modified sensitivity of
134 MEDICOGRAPHIA, VOL 31, No. 2, 2009
melatonin to the suppressant effect of light has been
demonstrated in bipolar patients and their offspring.21,22
The sleep-wake cycle is the most obvious circadian rhythm in humans, and sleep disturbances
represent a prominent feature of depression. Epidemiological studies estimate that 50% to 90% of
patients with diagnosed depression complain about
impairment of their sleep quality.23,24 Typically, the
complaints of depressed patients are about the difficulty in falling asleep, frequent nocturnal awakening, and early morning awakening. Insomnia is
thereby not only experienced subjectively, but also
reflected objectively in altered sleep architecture,
as first demonstrated by Kupfer and colleagues in
the early 1970s.25 These abnormalities consist of impaired sleep continuity and duration, a reduction of
slow-wave sleep (SWS), a shortening latency of the
initial rapid eye movement (REM) phase, an increase in the proportion of REM sleep in the early
part of the night, a prolongation of the first REM
period, an increased amount of total REM sleep,
and an increased number of eye movements during
REM periods (REM density).26 Longitudinal electroencephalogram studies in depressed patients have
described a tendency for REM sleep abnormalities to
resolve with improvement of the depression,59,60 and
even total normalization has been reported after
successful treatment.61 However, other studies have
reported the persistence of REM sleep and SWS abnormalities during remission, even after nonpharmacological treatments.62-64 Persistent and/or residual sleep disturbance has thereby been associated
with an increased risk of relapse,65 and the persistence of reduced SWS has similarly been associated with more rapid and more frequent recurrence
of depression.66
Motor activity
Motor activity shows a typical circadian rhythm in
humans, and most although not all investigations
have documented a phase advance (early daily peak)
of the circadian motor activity rhythm in bipolar
disorder patients in both the depressive and the
manic phases as well as during euthymia.27-29
Seasonal affective disorder
Seasonal changes in mood, appetite, sleep, and daily living function occur physiologically in many individuals. If these variations are of sufficient severity
to meet the criteria for a major depressive episode,
occur regularly during fall/winter, and are generally followed by a remission during the subsequent
spring and summer period, they may be regarded as
an episode of seasonal affective disorder (SAD). SAD
is a disorder with a circannual period, and patients
with SAD present with apparent chronobiological
abnormalities; hence, it is currently assumed that
SAD is a disorder of seasonal biological rhythms.67
Abnormalities of circadian rhythms in SAD patients
include sleep disturbances, quantitative changes
and phase delays in cortisol and melatonin secretion patterns, and increases in the minima of the
nocturnal body temperature as well as a phase delay of its 24-hour rhythm.68-70
Circadian rhythm disturbances in depression: remission quality/treatment implications – Monteleone and Maj
TRUE REMISSION
Disrupted circadian rhythms and
the pathogenesis of major depression
The fact that a wide variety of endogenous rhythms
are disrupted in individuals with depression has
led to speculation that such disturbances are not
unique to specific rhythms, but are associated, instead, with a disruption in the activity of the circadian master pacemaker in the SCN. Therefore, it is
plausible that alterations of the molecular components of the endogenous clock system play a role
in the disturbed circadian rhythms of mood disorder patients. The cellular machinery behind the circadian timing within the SCN neurons has been
largely identified, and it is believed to be under genetic control. Genes encoding essential elements of
the clock include, in mammals, period (per1, per2,
per3), neuronal PAS domain protein-2 (NPAS2),
circadian locomotor output cycles kaput (CLOCK),
cryptochrome (Cry1, Cry2), and brain and muscle
ARNT-like-1 (bmal1) genes. The proteins encoded
by these genes are part of a circadian autoregulatory loop incorporating activators and suppressors
of genes, whose activity thereby oscillates with a
circadian period, thus generating the endogenous
rhythmicity of SCN neurons.71
Both animal and human studies have provided
preliminary evidence of a role for circadian genes
in mood disorders. Mice carrying a mutation in the
CLOCK gene display a behavioral profile that is
strikingly similar to human mania, including hyperactivity, decreased sleep, reduced depression-like
behavior, lower anxiety, and an increase in the reward value for cocaine, sucrose, and medial forebrain bundle stimulation.72 Interestingly, many of
those mania-like behaviors are reverted by chronic
lithium administration and are rescued by expressing a functional CLOCK protein specifically in the
ventral tegmental area of CLOCK mutant mice.72
Studies in humans have begun to identify polymorphisms in certain circadian genes that are associated with mood disorders and, in particular,
bipolar disorder. The T3111C single nucleotide polymorphism (SNP) of the CLOCK gene has been investigated in both major depression and bipolar disorder. Whereas no differences were found in allelic
frequencies between individuals with a history of
major depression and healthy controls,30 the CC
genotype has been associated with a greater severity of insomnia during antidepressant treatment, a
higher recurrence rate of bipolar episodes, and a
reduced need for sleep in bipolar patients.31-33 In a
family-based sample of bipolar patients, an analysis
of 46 SNPs in 8 clock genes revealed a significant
although modest association of BMAL1 and TIM
genes with the mood disorder.34 An independent
study using haplotype analysis confirmed the association of bipolar disorder with the BMAL1 gene
and detected a new association with the PER3 gene.35
Finally, bipolar patients with the TT genotype of the
T50C SNP of the glycogen synthase kinase-3 gene,
which encodes an enzymatic protein regulating
central clock mechanisms, were found to be of an
earlier age at the onset of bipolar disorder and to experience less improvement from lithium therapy
IN
DEPRESSION: T
HE
ULTIMATE GOAL
than patients with the TC or CC genotypes.36,37 Recent studies suggest that SNPs of PER2, NPAS2,
and BMAL1 genes are associated with an increased
risk for SAD; furthermore, certain allelic combinations of SNPs of these three genes have an additive
effect, increasing the risk of developing SAD by 4.43
over other genotypes, and 10.67 over the most protective genotype.38
Based on the above findings, it could be suggested that primary or secondary alterations of the biological clock at the molecular level could lead to disruptions in endogenous circadian rhythms, which
in turn may generate the depressed state. Alternatively, it has been proposed that instead of or in addition to molecular abnormalities of the endogenous pacemaker, disturbances in environmental
zeitgebers may cause depressive symptoms in biologically predisposed individuals.73 This social zeitgeber theory specifically postulates that depressive
episodes arise as a consequence of life events causing a disturbance of social zeitgebers (ie, social factors such as the timing of meals, work schedules,
social demands, personal relationships), which, in
turn, derail an individual’s social rhythms. These disruptions can place substantial stress on the body’s
capacity to maintain stable biological rhythms, particularly sleep-wake, energy, alertness, and appetite
rhythms. Whereas in most individuals such rhythms
will restabilize shortly after the destabilizing events,
in predisposed subjects, they may precipitate a major depressive episode.
Finally, as suggested by Turek,74 the expression
of most rhythms at the behavioral, physiological,
and biochemical level is regulated by the integration
of inputs from the circadian clock and the sleepwake state of the organism. Thus, the circadian and
sleep control centers have evolved together to ensure a timely coordination between the internal and
external environment in order to optimize the survival of the species. Therefore, it could be that a primary circadian disturbance of the sleep-wake cycle
leads to insomnia that may desynchronize many endogenous rhythms, which then, in turn, may lead to
a depressed state. In support of this latter view, evidence has been provided that insomnia is a risk factor for the development of depression,75-77 as well as
for relapse and recurrence.78-80 Most of the circadian
abnormalities observed in the depressed state normalize with recovery, therefore it cannot be excluded that they arise as consequence of depression and
do not represent the primary determinants of the
affective disorder. However, even if so, the presence
of disrupted endogenous rhythms might potentially contribute to the maintenance of depressive
symptoms and might affect the course and/or the
prognosis of the affective episode. Therefore, circadian abnormalities of depressed patients are worthy
of clinical and therapeutic consideration.
Disrupted circadian rhythms
and the treatment of depression
The ideal antidepressant treatment should combine
high short-term efficacy for the acute phase of treatment with long-term efficacy and tolerability for
Circadian rhythm disturbances in depression: remission quality/treatment implications – Monteleone and Maj
MEDICOGRAPHIA, VOL 31, No. 2, 2009 135
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
the maintenance phase. This would result in a high
quality of remission, in which patients are asymptomatic with no or only minimal residual symptoms, and experience a full restoration of day-today functioning and quality of life. Currently used
antidepressant drugs, which act more or less specifically on brain monoamines, are frequently associated with significant limitations such as low remission rates, high risk of relapse, slow onset of
response, discontinuation symptoms, and side effects—especially sleep disturbances. Since a disruption of the normal circadian rhythmicity occurs at
least in a subgroup of depressed patients and is believed to play a role in the pathophysiology of depression, it is theoretically likely that interventions
able to induce phase shift within the circadian system, so that normal rhythmic patterns are restored,
may result in a high quality of remission. Therefore, chronotherapeutic interventions have been
developed, and these include both nonpharmacological strategies, such as sleep deprivation, light
therapy, and interpersonal and social rhythm therapy (IPSRT), and pharmacological treatments based
on the use of drugs specifically endowed with chronobiotic properties.
Sleep deprivation
One night of total sleep deprivation induces rapid
and effective, although short-lasting, antidepressant effects.39 Variants of total sleep deprivation,
such as selective REM sleep deprivation and partial
sleep deprivation, especially in the second half of
the night, are also effective, although total sleep
deprivation seems to be superior.39 However, the
therapeutic effect of sleep deprivation does not last
longer than one or maximally a few days, and this
intervention seems to work in less than 50% of patients.39
Light therapy
Light therapy is the treatment of choice for SAD,
where its astonishing success has led to the conclusion that it has to be considered the most successful clinical application of the circadian rhythm
concept in psychiatry. It moves from the hypothesis that reduced ambient light during fall/winter
leads to SAD symptoms in predisposed individuals40;
thus lengthening the photoperiod by exposing patients to bright light early in the morning before
dawn or in the evening has been proven to exert
antidepressant effects.41 It has subsequently been
proposed that most patients with SAD become depressed in fall/winter at least in part because the
later dawn in winter causes a delay in the patients’
endogenous circadian rhythms with respect to clock
time and the sleep-wake cycle.42 Therefore, providing a corrective phase advance should be useful in
realigning endogenous rhythms with the sleepwake cycle. In SAD patients, exposure to bright light
in the morning, which causes a phase advance of
endogenous circadian rhythms, has been shown to
produce higher antidepressant effects than exposure in the evening, which causes a phase-delay of
endogenous rhythms.40 However, some SAD patients
are actually phase-advanced, and this may explain
136 MEDICOGRAPHIA, VOL 31, No. 2, 2009
why in some studies, bright light scheduled in the
evening has been proven to have an antidepressant
effect that is equal to that of morning exposure.44
Guidelines for the treatment of SAD patients with
bright light have recently been provided.45 Standard
light treatment involves exposure to 1 to 2 hours
of a 2500-10000 lux light box in the morning immediately upon awakening, and for those patients
who do not respond to this schedule, a trial of
evening bright light (7-9 PM) may be necessary.
Interpersonal and social rhythm therapy
IPSRT was specifically designed to maintain regular daily rhythms, as well as identify and manage
potential precipitants of rhythm disruptions, in
accordance with the social zeitgeber theory that
depressive episodes arise as a consequence of life
events that disturb social zeitgebers.73 Therefore,
restoring the depressed patient’s social zeitgebers,
such as personal relationships, meals, exercise, and
social demands, would result in normalization of
biological rhythms and an improvement in mood.
Two preliminary studies have shown that although
increasing bipolar individuals’ social rhythm regularity did not improve their mood, participants
treated with IPSRT experienced longer episode-free
periods and were more likely to remain well in the
2-year preventive maintenance study phase.46,47
Agomelatine
As for pharmacologic interventions, melatonin has
been identified as having chronobiotic properties
in both rodents and humans81,82; therefore, it is supposed that the pineal hormone has an antidepressant effect. However, the few studies in which melatonin has been administered to depressed patients
have found an improvement in sleep, but no effect
on depressive symptoms83,84 and no enhancing effect on existing antidepressant therapies in patients
with treatment-resistant depression.85 By contrast,
agomelatine, a compound with agonistic properties at melatonergic MT1 and MT2 receptors and antagonistic properties at 5-HT2C receptors,86 which
are highly expressed in the SCN,87 has shown antidepressant properties in both preclinical and clinical
studies.88-91 It must be pointed out that the antidepressant effect of agomelatine is not solely mediated via its melatonergic action at the MT1 and MT2
receptors, but rather also depends on the compound’s 5-HT2C antagonistic property.92 This may
explain the above reported lack of antidepressant
action of exogenous melatonin, which acts only on
MT1 and MT2 receptors. It seems that, differently
from currently available antidepressant drugs, agomelatine possesses specific chronobiotic properties,
since it was able to regulate the sleep-wake cycle
and restore abnormal circadian rhythms in animal
models of disrupted circadian rhythms.48-50 In patients suffering from major depression, a recent
polysomnographic study indicated that agomelatine increased the duration of SWS and normalized
its distribution throughout the night.51 In a headto-head comparison study between venlafaxine and
agomelatine in major depressive disorder patients,
an earlier and better improvement of subjective
Circadian rhythm disturbances in depression: remission quality/treatment implications – Monteleone and Maj
TRUE REMISSION
IN
DEPRESSION: T
HE
Although a number of effective antidepressant drugs
have been introduced in recent years, there remain
significant unmet needs in the treatment of depression. Only approximately 30% of depressed patients
achieve remission, and even for remitted patients,
residual symptoms or drug side effects (eg, sleep disturbances, sexual dysfunction, weight gain) can re-
duce the quality of remission. Moreover, some antidepressant side effects can impair short-term and
long-term patient adherence to treatment, thus favoring no response, relapse, and/or recurrence.
At present, it is clear at the descriptive level that
some individuals with depression have circadian
rhythm abnormalities; whether there is, however,
a causal link between endogenous rhythm disruption and depression has not been firmly demonstrated, although evidence seems to be emerging
that this is the case. Nonetheless, improvement in
some forms of depression in response to strategies
that manipulate circadian rhythms support the idea
that circadian abnormalities observed in depressed
patients may constitute a core component of the
pathophysiology of depression. Therefore, normalization of circadian rhythms seems to represent a
possible new direction for the development of either pharmacological or nonpharmacological innovative therapeutic strategies to treat depression.
This new direction, in which circadian rhythms are
directly targeted, could hold promise for the identification of chronotherapeutic strategies that, compared with currently-available antidepressant treatments, could have more solid efficacy and fewer
side effects, achieving a better quality of remission
and a persistent amelioration of patients’ social functioning with a more complete restoration of their
quality of life. REFERENCES
1. Hastings MH. Central clocking. Trends Neurosci.1997;20:459464.
2. Welsh DK, Logothetis DE, Meister M, Reppert SM. Individual
neurons dissociated from rat suprachiasmatic nucleus express
independently phased circadian firing rhythms. Neuron. 1995;
14:697-706.
3. Moore RY, Speh JC. Serotonin innervation of the primate
suprachiasmatic nucleus. Brain Res. 2004;1010:169-173.
4. Wirz-Justice A. Biological rhythm disturbances in mood disorders. Int Clin Psychopharmacol. 2006;(suppl I):11-15.
5. Simonneaux V, Ribelayaga C. Generation of the melatonin endocrine message in mammals: a review of the complex regulation
of melatonin synthesis by norepinephrine, peptides, and other
pineal transmitters. Pharmacol Rev. 2003;55:325-395.
6. Buijs RM, Kalsbeek A. Hypothalamic integration of central and
peripheral clocks. Nat Rev Neurosci. 2001;2:521-526.
7. Sack RL, Auckley D, Auger RR, et al. Circadian rhythm sleep
disorders: part I, basic principles, shift work and jet lag disorders.
An American Academy of Sleep Medicine review. Sleep. 2007;30:
1460-1483.
8. Scott AJ. Shift work and health. Prim Care.2000;27:1057-1079.
9. Turner J, Drummond LM, Mukhopadhyay S, et al. A prospective
study of delayed sleep phase syndrome in patients with severe
resistant obsessive-compulsive disorder. World Psychiatry. 2007;
6:108-111.
10. Van Cauter E, Turek FW. Depression: a disorder of timekeeping? Perspect Biol Med. 1996;29:510-519.
11. Morris DW, Rush AJ, Jain S, et al. Diurnal mood variation
in outpatient with major depressive disorder: implications for
DSM-V from an analysis of the Sequenced Treatment Alternatives
to Relieve Depression Study data. J Clin Psychiatry. 2007;68:
1339-1347.
12. Germain A, Nofzinger EA, Meltzer CC, et al. Diurnal variation
in regional brain glucose metabolism in depression. Biol Psychiatry. 2007;62:438-445.
13. Souétre E, Salvati E, Wehr TA, Sack DA, Krebs B, Darcourt G.
Twenty-four-hour profile of body temperature and plasma TSH in
bipolar patients during depression and during remission and in
normal control subjects. Am J Psychiatry.1988;145:1133-1137.
14. Dietzel M, Saletu B, Lesch OM, Sieghart W, Schjerve M. Light
treatment in depressive illness: polysomnographic, psychometric
and neuroendocrinological findings. Eur Neurol. 1986;25(suppl II):93-103.
15. Goetze U, Tölle R. Circadian rhythm of free urinary cortisol,
temperature and heart rate in endogenous depressives and under
antidepressant therapy. Neuropsychobiology. 1987;18:175-184.
16. Parry BL, Mendelson WB, Duncan WC, Sack DA, Wehr TA.
Longitudinal sleep EEG, temperature, and activity measurements
across the menstrual cycle in patients with premenstrual depression and in age-matched controls. Psychiatry Res.1989;30:285303.
17. Souetre E, Salvati E, Belugou J-L, et al. Circadian rhythms in
depression and recovery: evidence for blunted amplitude as the
main chronobiological abnormality. Psychiatry Res.1989;28:263278.
18. Van Cauter E, Leprouolt R, Kupfer DJ. Effects of gender and
age on the levels and circadian rhythmicity of plasma cortisol.
J Clin Endocrinol Metab. 1996;81:2468-2473.
19. Claustrat B, Chazot G, Brun J, Jordan D, Sassolas G. A chronobiological study of melatonin and cortisol secretion in depressed
subjects: plasma melatonin, a biochemical marker in major depression. Biol Psychiatry. 1984;19:1215-1228.
20. Parry BL, Newton RP. Chronobiological basis of female-specific mood disorders. Neuropsychopharmacology. 2001;25(suppl V):102-108.
21. Lewy AJ, Nurnberger JI, Wehr TA, et al. Supersensitivity to
light: possible trait marker for manic-depressive illness. Am J
Psychiatry. 1985;142:725-727.
22. Nurnberger JI, Berrettini W, Tamarkin L, Hamovit J, Norton
J, Gershon E. Supersensitivity to melatonin suppression by light
in young people at high risk for affective disorder. A preliminary
report. Neuropsychopharmacology. 1988;1:217-223.
23. Casper RC, Redmond DE, Katz MM, Schaffer CB, Davis JM,
Koslow SH. Somatic symptoms in primary affective disorder:
presence and relationship to the classification of depression. Arch
Gen Psychiatry. 1985;42:1098-1104.
24. Riemann D, Berger M, Voderholzer U. Sleep and depression:
results from psychobiological studies: an overview. Biol Psychol.
2001;57:67-103.
25. Kupfer DJ, Foster FG. Interval between onset of sleep and
rapid-eye-movement sleep as an indicator of depression. Lancet.
1972;2:684-686.
26. Thase ME, Carpenter L, Kupfer DJ, Frank E. Clinical significance of reversed vegetative subtypes of major depression. Psychopharmacol Bull. 1991;27:17-22.
27. Foster FG, Kupfer DJ. Psychomotor activity as a correlate
of depression and sleep in acutely disturbed psychiatric inpatients.
Am J Psychiatry. 1975;132:928-931.
measures of getting to sleep, quality of sleep, and
ease of awakening have been reported with agomelatine.52 Moreover, agomelatine has been shown to
be significantly superior to placebo in rates of clinical response and remission in a study in which the
remission criteria (Hamilton Rating Scale for Depression score 6) were more stringent than usual53; the comparator in this study, paroxetine, had
lower response and remission rates.53 Finally, in all
the above clinical trials, agomelatine exhibited a
favorable sexual side-effect profile and was in general well tolerated. All these data are indicative of
agomelatine’s potential to renormalize circadian
rhythms, including sleep-wake cycle alterations,
without sedative effects and no sexual impairment,
which could possibly lead to better adherence to
antidepressant treatment, optimization of the
achievement of full recovery with a high quality remission, and restoration of patients’ quality of life.
Conclusion
Circadian rhythm disturbances in depression: remission quality/treatment implications – Monteleone and Maj
ULTIMATE GOAL
MEDICOGRAPHIA, VOL 31, No. 2, 2009 137
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
28. Wolff EA, Putnam FW, Post RM. Motor activity and affective
illness: relationship of amplitude and temporal distribution to
changes in affective states. Arch Gen Psychiatry.1985;42:288-294.
29. Salvatore P, Ghidini S, Zita G, et al. Circadian activity rhythm
abnormalities in ill and recovered bipolar I disorder patients.
Bipolar Disord. 2008;10:256-265.
30. Desan PH, Oren DA, Malison R, et al. Genetic polymorphism
at the CLOCK gene locus and major depression. Am J Med Genet.
2000;96:418-421.
31. Benedetti F, Serretti A, Colombo C, et al. Influence of CLOCK
gene polymorphism on circadian mood fluctuation and illness
recurrence in bipolar depression. Am J Med Genet. 2003;123:
23-26.
32. Serretti A, Benedetti F, Mandelli L, et al. Genetic dissection
of psychopathological symptoms: insomnia in mood disorders
and CLOCK gene polymorphism. Am J Med Genet. 2003;121:3538.
33. Serretti A, Cusin C, Benedetti F, et al. Insomnia improvement during antidepressant treatment and CLOCK gene polymorphism. Am J Med Genet. 2005;137:36-39.
34. Mansour HA, Wood J, Logue T, et al. Association study of eight
circadian genes with bipolar I disorder, schizoaffective disorder
and schizophrenia. Genes Brain Behav. 2006;5:150-157.
35. Nievergelt CM, Kripke DF, Barrett TB, et al. Suggestive evidence for association of the circadian genes PERIOD3 and ARNTL
with bipolar disorder. Am J Med Genet. 2006;141:234-241.
36. Benedetti F, Bernasconi A, Lorenzi C, et al. A single nucleotide
polymorphism in glycogen synthase kinase 3-beta promoter gene
influences onset of illness in patients affected by bipolar disorder.
Neurosci Lett. 2004;355:37-40.
37. Benedetti F, Serretti A, Pontiggia A, et al. Long-term response
to lithium salts in bipolar illness is influenced by the glycogen
synthase kinase 3-beta -50 T/C SNP. Neurosci Lett. 2005;376:5155.
38. Partonen T, Treutlein J, Alpman A, et al. Three circadian clock
genes Per2, Arntl, and Npas2 contribute to winter depression.
Ann Med. 2005;39:229-238.
39. Giedke H, Schwarzler F. Therapeutic use of sleep deprivation
in depression. Sleep Med Rev. 2002;6:361-377.
40. Rosen LN, Targum SD, Terman M, et al. Prevalence of seasonal
affective disorder at four latitudes. Psychiatry Res. 1990;31:131144.
41. Wehr TA, Jacobsen FM, Sack DA, Arendt J, Tamarkin L, Rosenthal NE. Phototherapy of seasonal affective disorder. Time of day
and suppression of melatonin are not critical for antidepressant
effects. Arch Gen Psychiatry. 1986;43:870-875.
42. Lewy AJ, Lefler BJ, Emens JF, Bauer VK. The circadian basis
of winter depression. Proc Natl Acad Sci U S A. 2006;103:74147419.
43. Golden RN, Gaynes BN, Ekstrom RD, et al. The efficacy of
light therapy in the treatment of mood disorders: a review and
meta-analysis of the evidence. Am J Psychiatry. 2005;162:656662.
44. Wirz-Justice A, Graw P, Krauchi K, et al. Light therapy in seasonal affective disorder is independent of time of day or circadian phase. Arch Gen Psychiatry. 1993;50:929-937.
45. Lewy AJ, Rough JN, Songer JB, Mishra N, Yuhas K, Emens JS.
The phase shift hypothesis for the circadian component of winter
depression. Dialogues Clin Neurosci. 2007;9:291-299.
46. Frank E, Hlastala S, Ritenour A, et al. Inducing lifestyle regularity in recovering bipolar disorder patients: results from the
maintenance therapies in bipolar disorder protocol. Biol Psychiatry. 1997;41:1165-1173.
47. Frank E, Kupfer DJ, Thase ME, et al. Two-year outcomes for
interpersonal and social rhythm therapy in individuals with bipolar I disorder. Arch Gen Psychiatry. 2005;62:996-1004.
48. Martinet L, Guardiola-Lemaitre B, Mocaer E. Entrainment of
circadian rhythms by S-20098, a melatonin agonist, is dose and
plasma concentration dependent. Pharmacol Biochem Behav.
1996;54:713-718.
49. Armstrong SM, McNulty OM, Guardiola-Lemaitre B, Redman
JR. Successful use of S20098 and melatonin in an animal model
of delayed sleep-phase syndrome (DSPS). Pharmacol Biochem
Behav. 1993;46:45-49.
50. Fuchs E, Schmelting B, Mocaer E. Effects of the novel antidepressant agomelatine (S 20098) and fluoxetine in chronically
stressed tree shrews, an animal model of depression. Eur Neuropsychopharmacol. 2006;16(suppl IV)IV338.
51. Quera Salva MA, Vanier B, Laredo J, et al. Major depressive
disorder, sleep EEG and agomelatine: an open-label study. Int J
Neuropsychopharmacol. 2007;10:691-696.
52. Lemoine P, Guilleminault C, Alvarez E. Improvement in subjective sleep in major depressive disorder with a novel antidepressant, agomelatine: randomized, double-blind comparison with
138 MEDICOGRAPHIA, VOL 31, No. 2, 2009
venlafaxine. J Clin Psychiatry. 2007;68:1723-1732.
53. Loo H, Hale A, D’haenen H. Determination of the dose of agomelatine, a melatonergic agonist and a selective 5-HT(2C) antagonist, in the treatment of major depressive disorder: a placebocontrolled dose range study. Int Clin Psychopharmacol. 2002;17:
239-247.
54. Leibenluft E, Nooman BM, Wehr TA. Diurnal variation: reliability of measurement and relationship to typical and atypical
symptoms of depression. J Affect Disord. 1992;26:199-204.
55. Peeters F, Berkhof J, Delenspaul P, Rottenberg J, Nicolson NA.
Diurnal mood variation in major depressive disorders. Emotion.
2006;6:383-391.
56. Duncan WC. Circadian rhythms and the pharmacology of affective illness. Pharmacol Ther. 1996;71:253-312.
57. Avery D, Wildschiodtz G, Rafaelsen O. REM latency and temperature in affective disorder before and after treatment. Biol Psychiatry. 1982;17:463-470.
58. Carvalho LA, Gorenstein C, Moreno RA, Markus RP. Melatonin
levels in drug-free patients with major depression from the southern hemisphere. Psychoneuroendocrinology. 2006;31:761-768.
59. Cartwright RD. Rapid eye movement sleep characteristics
during and after mood-disturbing events. Arch Gen Psychiatry.
1983;40:197-201.
60. Buysse DJ, Frank E, Lowe KK, Cherry CR, Kupfer DJ. Electroencephalographic sleep correlates of episode and vulnerability to
recurrence in depression. Biol Psychiatry. 1997;41:406-418.
61. Knowles JB, Cairns J, McLean AW, et al. The sleep of remitted
bipolar depressives: comparison with sex and age-matched controls. Can J Psychiatry. 1986;31:295-298.
62. Rush AJ, Erman MK, Giles DE, et al. Polysomnographic findings in recently drug-free and clinically remitted depressed patients. Arch Gen Psychiatry. 1986;43:878-884.
63. Giles DE, Etzel BA, Reynolds CF, Kupfer DJ. Stability of polysomnographic parameters in unipolar depression: a cross-sectional report. Biol Psychiatry. 1986;25:807-810.
64. Thase ME, Fasiczka AL, Berman SR, Simons AD, Reynolds CF.
Electroencephalographic sleep profiles before and after cognitive
behaviour therapy of depression. Arch Gen Psychiatry.1998;55:
138-144.
65. Giles DE, Jarrett RB, Roffwarg HP, Rush AJ. Reduced rapid
eye movement latency: a predictor of recurrence in depression.
Neuropsychopharmacology. 1987;1:33-39.
66. Kupfer DJ, Frank E, McEachran AB, Grochocinski VJ. Delta
sleep ratio: a biological correlate of early recurrence in unipolar
affective disorder. Arch Gen Psychiatry. 1990;47:1100-1105.
67. Levitan RD. The chronobiology and neurobiology of winter
seasonal affective disorder. Dialogues Clin Neurosci.2007;9:315324.
68. Dahl K, Avery DH, Lewy AJ, et al. Dim light melatonin onset
and circadian temperature during a constant routine in hypersomnic winter depression. Acta Psychiatr Scand.1993;88:60-66.
69. Avery DH, Dahl K, Savage MV, et al. Circadian temperature
and cortisol rhythms during a constant routine are phase-delayed
in hypersomnic winter depression. Biol Psychiatry. 1997;41:
1109-1123.
70. Schwartz PJ, Rosenthal NE, Turner EH, Drake CL, Liberty V,
Wehr TA. Seasonal variation in core temperature regulation during sleep in patients with winter seasonal affective disorder. Biol
Psychiatry. 1997;42:122-131.
71. Laposky AD, Shelton J, Bass J, Dugovic C, Perrino N, Turek FW.
Altered sleep regulation in leptin deficient mouse. Am J Physiol
Regul Integr Comp Physiol. 2006;290:R894-R903.
72. Roybal K, Theobold D, Graham A, et al. Mania-like behavior
induced by disruption of CLOCK. Proc Natl Acad Sci U S A. 2007;
104:6406-6411.
73. Ehlers CL, Frank E, Kupfer DJ. Social zeitgebers and biological rhythms. Arch Gen Psychiatry. 1988;45:948-952.
74. Turek FW. From circadian rhythms to clock genes in depression. Int Clin Psychopharmacol. 2007;22(suppl II):1-8.
75. Ford DJ, Kamerow DB. Epidemiological study of sleep disturbances and psychiatric disorders. Am J Med Assoc. 1989;262:
1479-1484.
76. Eaton WW, Badawi M, Melton B. Prodromes and precursors:
epidemiological data for primary prevention of disorders with
slow onset. Am J Psychiatry. 1995;152:967-972.
77. Chang PP, Ford DE, Mead LA, Cooper-Patrick L, Klag MJ. Insomnia in young men and subsequent depression. The John Hopkins precursors study. Am J Epidemiol. 1997;146:105-114.
78. Armitage R. The effects of antidepressants on sleep in patients
with depression. Can J Psychiatry. 2000;45:803-809.
79. Ohayon MM, Roth T. Place of chronic insomnia in the course
of depressive and anxiety disorders. J Psychiatr Res. 2003;37:9-15.
80. Dombrovski AY, Mulsant BH, Houck PR, et al. Residual symptoms and recurrence during maintenance treatment of late-life
Circadian rhythm disturbances in depression: remission quality/treatment implications – Monteleone and Maj
TRUE REMISSION
depression. J Affect Disord. 2007;103:77-82.
81. Van Reeth O, Olivares W, Turek FW, et al. Comparative effects
of a melatonin-agonist on the circadian system in mice and Syrian hamster. Brain Res. 1997;762:185-194.
82. Lewy AJ, Sack RL. Exogenous melatonin’s phase-shifting effects on the endogenous melatonin profile in sighted humans: a
brief review and critique of the literature. J Biol Rhythms.1997;
12:588-594.
83. de Wries MW, Peeters FP. Melatonin as a therapeutic agent in
the treatment of sleep disturbances in depression. J Nerv Ment
Dis. 1997;185:201-202.
84. Dolberg OT, Hirschmann S, Grunhaus L. Melatonin for the
treatment of sleep disturbances in major depressive disorder. Am
J Psychiatry. 1998;155:1119-1121.
85. Dalton EJ, Rotondi D, Levitan RD, Kennedy SH, Brown GM.
Use of slow-release melatonin in treatment-resistant depression.
J Psychiatry Neurosci. 2000;25: 48-52.
86. Millan MJ, Gobert A, Lejeune F, et al. The novel melatonin
agonist agomelatine (S20098) is an antagonist at 5-hydroxytryptamine2C receptors, blockade of which enhances the activity of
frontocortical dopaminergic and adrenergic pathways. J Pharmacol Exp Ther. 2003;306:954-964.
87. Liu C, Weaver DR, Jin X, et al. Molecular dissection of two
IN
DEPRESSION: T
HE
ULTIMATE GOAL
distinct actions of melatonin on the suprachiasmatic circadian
clock. Neuron. 1997;19:91-102.
88. Loo H, Daléry J, Macher JP, Payen A. Pilot study comparing
in blind the therapeutic effect of two doses of agomelatine, melatonin- agonist and selective 5HT2c receptors antagonist, in the
treatment of major depressive disorders. Encephale. 2003;29:
165-171.
89. Bourin M, Mocaër E, Porsolt R. Antidepressant-like activity
of S 20098 (agomelatine) in the forced swimming test in rodents:
involvement of melatonin and serotonin receptors. J Psychiatry
Neurosci. 2004;29:126-133.
90. Barden N, Shink E, Labbé M, Vacher R, Rochford J, Mocaër E.
Antidepressant action of agomelatine (S 20098) in a transgenic
mouse model. Prog Neuropsychopharmacol Biol Psychiatry.
2005;29:908-916.
91. Olié JP, Kasper S. Efficacy of agomelatine, a MT1/MT2 receptor agonist with 5-HT2C antagonistic properties, in major depressive disorder. Int J Neuropsychopharmacol. 2007;10:661673.
92. Gruca P, Przegalinski E, Mrowiec S, Lason M, Papp M. Evidence for antidepressant- and anxiolytic-like activities of melatonin and agomelatine in animal models. Eur Neuropsychopharmacol. 2004;14:230.
TROUBLES
DU RYTHME CIRCADIEN DANS LA DÉPRESSION
CONSÉQUENCES POUR LE TRAITEMENT ET
LA QUALITÉ DE LA RÉMISSION
:
L
es troubles de l’humeur, et en particulier les troubles dépressifs unipolaires et les troubles affectifs saisonniers, sont liés à des anomalies endogènes du rythme circadien. La perturbation de la rythmicité circadienne
normale interviendrait au moins dans un certain sous-groupe de patients déprimés, le lien causal entre la perturbation du rythme circadien endogène et la
dépression n’ayant néanmoins pas été formellement démontré. Cependant,
l’amélioration de certaines formes de dépression en réponse à des stratégies
agissant sur les rythmes circadiens permet de penser que les anomalies circadiennes observées chez les patients déprimés pourraient constituer une composante clé de la physiopathologie de la dépression méritant d’être prise en considération. Des actions chronothérapeutiques, comprenant à la fois des stratégies
non pharmacologiques, telles que la privation de sommeil, la luminothérapie
et la psychothérapie interpersonnelle et des rythmes sociaux (PTIRS), ainsi que
des traitements pharmacologiques fondés sur l’utilisation de médicaments dotés de propriétés chronobiotiques, comme l’agomélatine, ont montré des effets
antidépresseurs efficaces. Ainsi, la normalisation des rythmes circadiens représente une nouvelle direction possible pour le développement de stratégies thérapeutiques innovantes, pharmacologiques ou non, qui pourraient avoir un
rôle futur important comme alternative ou adjuvant aux traitements antidépresseurs actuellement disponibles, afin d’obtenir une meilleure qualité de rémission et une amélioration durable de la qualité de vie et de fonctionnement
social des patients.
Circadian rhythm disturbances in depression: remission quality/treatment implications – Monteleone and Maj
MEDICOGRAPHIA, VOL 31, No. 2, 2009 139
IN
DEPRESSION: THE ULTIMATE GOAL
TRUE REMISSION
Constantin R. SOLDATOS,* MD
Christos G. THELERITIS,*† MD
*National and Kapodistrian University of
Athens, Mental Health Care Unit Evgenidion
Hospital, Athens, GREECE
†National and Kapodistrian University of
Athens, First Psychiatry Department
Eginition Hospital, Athens
GREECE
Improvement of the
sleep-wake cycle as a target
for remission in depression
by C. R. Soldatos and
C. G. Theleritis, Greece
Sleep in depression
t has long been reported that sleep is disturbed
in depression.1-5 Epidemiological studies have
demonstrated that 50% to 90% of patients suffering from depression complain of poor sleep.6 In
a large European community study, for instance,
63% of patients diagnosed with depression reported sleep problems.7 On the other hand, it has been
reported that about 20% of subjects with insomnia
screened in general population studies are found to
be depressed.8-10 Moreover, subjective reports of sleep
disturbance and objectively assessed polysomnographic abnormalities have been correlated with an
increased risk of new-onset major depression,11 persistence of depressive symptomatology,12 and recur-
I
leep disorders have been long associated with depression; they are often
used to identify newly presenting depressive patients, and may be part of
a more general alteration of biological rhythms. Furthermore, persistent
insomnia is a common residual symptom in incompletely remitted depression.
As a rule, to avoid the recurrence of depressive symptomatology, treatment of
associated insomnia should be pursued in patients taking antidepressants. Combining pharmacotherapy with nonpharmacological treatments that take into
account the sleep-wake cycle (sleep deprivation, light therapy) could help in the
resolution of both depressive symptoms and associated sleep disorders. A novel
pharmaceutical agent, agomelatine, an agonist of melatonergic MT1 and MT2
receptors, as well as an antagonist of 5-HT2C receptors, shows a unique synchronizing effect on circadian rhythms and demonstrates robust antidepressant
efficacy.
S
Medicographia. 2009;31:140-145.
(see French abstract on page 145)
Keywords: insomnia; sleep disorder; slow-wave sleep; rapid eye movement;
incomplete remission; circadian rhythm; psychotherapy; pharmacotherapy
www.medicographia.com
Address for correspondence: Professor Constantin R. Soldatos, National and Kapodistrian University of
Athens, Mental Health Care Unit, Evgenidion Hospital, Athens 11528, Greece
(e-mail: [email protected])
140 MEDICOGRAPHIA, VOL 31, No. 2, 2009
rence of depression following successful treatment.13
Polysomnographic studies have documented several sleep disturbances in depression: difficulty in
initiating and maintaining sleep, prolonged sleep
onset latency, multiple awakenings during the night,
early morning awakening, and decreased total sleep
time.1,3,5,6 Furthermore, slow-wave sleep (SWS) and
rapid eye movement (REM) sleep are often found to
differ between depressed patients and healthy individuals.1,14 Abnormal sleep architecture in depressed
patients is reflected in reduced SWS,15 a greater
number of stage shifts, earlier onset and a higher
percentage of REM sleep, especially in the early part
of the night, and an increased REM density.16
It should also be noted that computerized sleep
electroencephalogram (EEG) analysis of SWS frequently reveals a lower level of delta wave activity
during the first non-REM period than in the second
non-REM period, ie, a lower delta sleep ratio, which
is contrary to what happens in normal individuals.17
However, it should be noted that no single sleep abnormality is specific for depression, because similar polysomnographic aberrations have also been
reported in several other psychiatric conditions.1
SELECTED
CBT
EEG
GABA
IPSRT
REM
SCN
SNRI
SSRI
SWS
TCA
ABBREVIATIONS AND ACRONYMS
cognitive behavioral therapy
electroencephalogram
gamma aminobutyric acid
interpersonal and social rhythm therapy
rapid eye movement
suprachiasmatic nucleus
serotonin and norepinephrine reuptake
inhibitor
selective serotonin reuptake inhibitor
slow-wave sleep
tricyclic antidepressant
Improvement of the sleep-wake cycle as a target for remission in depression – Soldatos and Theleritis
TRUE REMISSION
Mechanisms for sleep changes
in depression
Sleep mechanisms and the pathophysiology of depression are closely interrelated. Monoaminergic
and cholinergic neurotransmission are heavily involved in both.18 Therefore, it is not surprising that
depression is almost invariably associated with sleep
abnormalities. Several hypotheses have been proposed to explain their occurrence. The first hypothesis suggests that an imbalance between the monoaminergic and cholinergic systems in the central
nervous system could be responsible for both the
pathophysiology of depression and the observed
sleep aberrations.18-20 According to another hypothesis, increased pressure of REM sleep might be responsible18,21; however, a number of medications
(bupropion, nefazodone, and trazodone) with welldocumented antidepressant efficacy do not suppress
REM sleep, which suggests that REM sleep suppression might not be necessary for an antidepressant
treatment response.22 The third hypothesis proposes an hypothalamic-pituitary-adrenal axis dysregulation.23
The last hypothesis suggests that a deficiency in
the mechanism responsible for non-REM sleep, as
explained by the two-process model of sleep regulation, may be implicated. According to Borbély,24
the interaction of two processes is responsible for
sleep regulation: a homeostatic process (process S),
with an increase of EEG slow waves during waking
and an exponential decline during sleep, interacts
with a circadian process (process C)— an internal
clock—so that the timing and architecture of sleep
is determined. It was proposed that process S is deficient in patients with depression. In fact, the clinical sleep disturbance of early morning awakening
could be attributed to an impaired functioning of
process S during waking or an earlier timing of
process C. The fast antidepressant effect gained
from sleep deprivation might arise from an increase
in process S to normal levels.24 By contrast, the slow
antidepressant effect of a phase advance of the sleepwake cycle might be related to gradual shifts of process S toward a correct phase relationship with regard to process C. However, it should be noted that
the antidepressant effect of phase advance has not
been confirmed in all studies.25 Moreover, decline of
either process S during sleep or of the phase and
amplitude of process C are other possible abnormalities that might be involved in sleep disturbances.26
Circadian rhythms in depression
The biological clock in the suprachiasmatic nucleus (SCN), a master pacemaker driving circadian
rhythms in the brain and body, is synchronized to
the external light-dark cycle via retinal light input.
Nocturnal synthesis of the pineal hormone melatonin is driven by the SCN. Yet, circadian oscillators
are found in every organ,27 and each organ has its
own appropriate synchronizer. Although light is the
major synchronizer for the SCN, it does not affect
clocks in the liver; the synchronizer for the latter is
food, but food is not a synchronizer for the SCN.27
IN
DEPRESSION: T
HE
ULTIMATE GOAL
Individuals have different preferences for timing
their sleep28; some like to go to sleep early and wake
up early in the morning, while others go to sleep
late and wake up late. In addition, individual genetic characteristics of the molecular mechanisms
of the biological clock might determine features
of mood disorders, including age at onset,29 recurrence,30 symptoms of insomnia and response to its
treatment,31,32 and response to sleep deprivation.29
Furthermore, it has been proposed that there is an
intimate relationship between the neurotransmitter systems targeted by drugs and the circadian
rhythms targeted by chronotherapeutics.33 These
genetic factors may actually provide a chronobiological vulnerability for depression; in this case, a
“double desynchronization” may occur—“internal
desynchronization” between different clocks in the
body and brain, and “external desynchronization”
between the timing of body rhythms with respect
to the light-dark cycle.33
Stable internal and external phase relationships
appear to be crucial for a stable mood state (ie, the
timing between cortisol and temperature body
rhythms as well as the timing of sleep with respect to
the day-night cycle).34 Any desynchronization might
cause mood disturbances, particularly in vulnerable
individuals. Certain synchronizers have been used
to stabilize phase, with light and melatonin being
the most important, but also sleep deprivation having been successfully applied in everyday practice.35,36
Treatment considerations
Nonpharmacological interventions on
circadian rhythms
Manipulations of the sleep-wake cycle, whether its
duration (total or partial sleep deprivation) or its
timing (partial sleep deprivation, phase advance),
have profound and rapid effects on depressed mood
in 60% of all diagnostic subgroups of affective disorders.37 The therapeutic effect of sleep deprivation
is postulated to be linked to an increase in homeostatic sleep pressure; additionally, sleep deprivation–induced sleepiness may counteract the hyperarousal state that is often present in depression.26
Though sleep deprivation may have a transient
effect on mood, it should always be considered as
an option, since it might be the most rapid antidepressant therapeutic solution35,36; however, most patients tend to relapse soon after recovery of sleep.
The combination of sleep deprivation with lithium
(with or without light therapy)38-40 or pindolol41 may
prevent relapse.
Light therapy was specifically developed as a synchronizer in the treatment of patients with seasonal affective disorders42; however, it has also shown
efficacy in nonseasonal depression,43,44 and it can
prevent relapse after sleep deprivation.45 Bright light
has three major effects on the circadian system: it
increases circadian amplitude, shifts circadian phase
(depending on the time of its application), and
thereby modifies the phase relationships between
the internal clock and sleep, and the external lightdark cycle.26 As mentioned, it has been reported that
bright light can prevent relapse after sleep depriva-
Improvement of the sleep-wake cycle as a target for remission in depression – Soldatos and Theleritis
MEDICOGRAPHIA, VOL 31, No. 2, 2009 141
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
tion; it was also recently demonstrated that light
therapy combined with a selective serotonin reuptake inhibitor (SSRI) leads to more rapid and more
profound improvement in patients with nonseasonal major depression,45,46 suggesting an advantage of
using combined approaches.
Psychotherapeutic and psychosocial
interventions
There is evidence from studies of primary insomnia
that cognitive behavioral therapy (CBT) results in
improvements that are as substantial as those of
pharmacotherapy with sedative-hypnotics.47 The
greatest advantage of CBT is that its effectiveness is
more durable than that of pharmacotherapy and the
benefits persist after therapy is terminated.48 CBT
has also been reported to be efficient in the management of depression.49 Further research, however,
is needed regarding the efficacy of CBT—in combination with antidepressant therapy—for insomniac patients with major depressive disorder.50
Behavioral modification of social rhythms may
help in the treatment of depression. Frank and colleagues developed the Interpersonal and Social
Rhythm Therapy (IPSRT) Intervention Program in
order to establish regular social rhythm regularity
(having regular bed, wake, and meal times, switching to a more regular work schedule, and incorporate a regular daily exercise session). IPSRT reduces
the risk of recurrence in bipolar patients, regularizes
social rhythms, accelerates remission in depressed
patients, and lowers relapse rates; these effects are
comparable with those of intensive pharmacotherapy.51 Further studies should determine whether
IPSRT should be used alone or as an adjunctive
treatment in major depression.52
Psychopharmacological treatment approaches
Melatonin, exogenously administered, acts as a synchronizer of circadian rhythms and sleep (eg, in
blind persons) but does not appear to have any major effects on mood.53 Antidepressants alleviate symptoms of depression by altering the levels of various
central nervous system neurotransmitters that are
also involved in sleep regulation, and may have a
considerable impact on sleep patterns. In general,
the majority of antidepressant drugs suppress REM
sleep and increase REM latency, although this is not
always the case. As far as sleep efficiency and total
sleep time are concerned, antidepressants can be
distinguished as being either sedative or energizing.54 Currently-available antidepressants have notable limitations in terms of their only moderate efficacy compared with placebo, relatively slow onset
of action, possible withdrawal symptoms, and problems of compliance. Sleep disturbances are often
used to identify newly presenting depressive patients, and may be part of a more general alteration
of bodily rhythms.55 Persistent insomnia is one of
the most common residual symptoms in incompletely remitted depression.56
Residual depressive symptoms are one of the predictors of subsequent relapse risk,57,58 and of persistent functional disability. Complete relief of associated insomnia should be pursued in patients under
142 MEDICOGRAPHIA, VOL 31, No. 2, 2009
treatment with antidepressants to avoid the recurrence of depressive symptomatology.59 In general,
the sleep disturbances associated with depression
improve as depressive symptom severity lessens
with treatment, but different antidepressants have
characteristic effects on sleep, particularly in the
early stages of treatment.
On short-term administration, most tricyclic antidepressants (TCAs) promote sleep by increasing
total sleep time and SWS. Furthermore, REM sleep
suppression is reported with all TCAs except trimipramine.60,61 More specifically, amitriptyline, trimipramine, nortriptyline, dothiepin, and doxepin have
all been associated with sedation (although imipramine62 and desipramine63 are less likely to be linked
with sedation and have been associated with insomnia instead). Because of their sedative properties,
these agents are often used as hypnotics in depressed
patients suffering from insomnia.64-68 However, daytime drowsiness and poor functioning during the
next day may be an untoward side effect of sedative
TCAs, in particular amitriptyline and doxepin. It
should also be noted that TCAs may cause insomnia
by inducing or worsening periodic limb movement
disorder, and that their use is often associated with
nightmares.69
The reversible monoamine oxidase inhibitor moclobemide is associated with less REM sleep suppression and appears not to affect sleep notably.70
SSRIs are considered to be more energizing agents
than other antidepressants. Although SSRIs are
commonly associated with insomnia, sedation and
daytime sleepiness have been occasionally reported
with high doses of these drugs. Fluvoxamine and
paroxetine have been reported as being more sedating than other SSRIs.22,26,67,69 Objective polysomnographic findings on the effects of SSRIs on sleep
show prolongation of sleep onset latency, increased
wakefulness, decreased sleep efficiency, increased
number of arousals during the night, increased
REM latency, and decreased total REM sleep, when
compared with placebo and sedative TCAs.64-67,69
Furthermore, SSRIs can exacerbate periodic limb
movement disorder and can cause nightmares.
However, patients’ subjective sleep ratings whilst
taking SSRIs are frequently positive. The same is
also true in terms of clinicians’ ratings of sleep items
on the Hamilton Rating Scale for Depression in
clinical trials that investigated citalopram, sertraline, fluoxetine, fluvoxamine, and paroxetine. However, it is unlikely that a depressed patient with a
history of severe sleep disturbance will benefit more
from SSRI treatment than from the administration
of more sedative antidepressants.
Venlafaxine,70,71 a serotonin and norepinephrine
reuptake inhibitor (SNRI), decreases total sleep time
and may cause insomnia in 4% to 18% of depressed
patients. Also, its administration has been associated with REM sleep behavior disorder. Venlafaxine
and duloxetine72 (another SNRI), as well as reboxetine, a norepinephrine reuptake inhibitor, appear
to have a similar profile to SSRIs regarding their
influence on REM sleep and overall sleep architecture. Another antidepressant that may cause insomnia is bupropion, a dopamine and norepinephrine
Improvement of the sleep-wake cycle as a target for remission in depression – Soldatos and Theleritis
TRUE REMISSION
reuptake inhibitor, which also decreases REM sleep
and lengthens REM latency.
Trazodone, a serotonin, α1-adrenergic, and histamine receptor blocker, increases total sleep time,
enhances SWS, and may cause excessive daytime
sleepiness. Trazodone does not suppress REM sleep.5
It is a very sedative antidepressant and has often
been used for the management of severe insomnia
in depression. A number of trials have shown that
trazodone is more effective than the TCAs and
equally as effective as a hypnotic agent, zolpidem,
in improving sleep.64-67,69 Nefazodone, a compound
closely related to trazodone, is a potent antagonist
of postsynaptic 5-HT2 receptors, but with a less potent α1-adrenoreceptor–blocking activity and no
affinity for histamine receptors. It promotes sleep
without suppressing REM sleep.5 Its pharmacological profile makes it one of the best antidepressants
to relieve insomnia in the context of depression, and
it is widely used as an adjunct to SSRIs and SNRIs.5
Also, maprotiline, which acts by inhibiting reuptake of norepinephine and blocking the histamine
receptor, causes sedation and suppresses REM sleep.
Similarly, amoxapine, which blocks the serotonin
and histamine receptors and inhibits the uptake
of norepinephrine, is another antidepressant that
causes sedation. Tianeptine, a glutamate modulator,73 enhances sleep continuity and ameliorates
subjective sleep quality without significantly modifying REM sleep parameters.54
Mirtazapine is a selective serotonin and α2adrenoreceptor blocker, which also blocks histamine
receptors (noradrenergic and specific serotonergic
antidepressant). Potent antagonism of H1 receptors
causes immediate, nonspecific sedative effects upon
administration. Mirtazapine increases total sleep
time and suppresses REM sleep. It is frequently used
when sleep complaints are a major concern in depression and its efficacy is comparable to that of
sedative TCAs.64-67,69
Agomelatine, an agonist of melatonergic MT1 and
MT2 receptors, as well as an antagonist of 5-HT2C
receptors, uniquely combines circadian synchronizing effects with selective norepinephrine and
dopamine augmentation properties in the prefrontal
cortex, and demonstrates robust antidepressant efficacy.74-76 When administered in the evening, it advances circadian phase and directly increases sleepiness through thermoregulatory mechanisms.77,78
This promotes a rapid sleep onset, but is without
any after-effects the following day. The drug does
not raise extracellular serotonin levels, and that is
why it does not produce any of the side effects (diarrhea, nausea, sexual dysfunction, insomnia) usually observed with SSRIs. Furthermore, agomelatine does not block histaminergic, α1-adrenergic,
and muscarinic receptors, and as a result does not
share the side effects of TCAs.79
Combination therapy
There is evidence from everyday clinical experience,
and a few controlled studies as well, that combining
benzodiazepine hypnotics80 or nonbenzodiazepine
selective gamma aminobutyric acid (GABA) type A
receptor antagonists, such as zolpidem, zopiclone,
IN
DEPRESSION: T
HE
ULTIMATE GOAL
eszopiclone, and zaleplon, with antidepressants
from the beginning of therapy may result in a faster
relief of the concomitant sleep disturbance, and
thereafter bring about a faster improvement in depressive symptoms.81 Therefore, it is common practice to use such combinations for the initial management of insomnia in the context of depression.5
Benzodiazepines commonly used for the management of insomnia are lorazepam, temazepam, alprazolam, oxazepam, triazolam, diazepam, flurazepam, and midazolam. All benzodiazepines shorten
sleep onset latency, increase total sleep time and
stage 2 sleep, and suppress REM sleep and SWS. Excessive daytime sleepiness is a troublesome side effect, which is more pronounced with long-acting
benzodiazepines. Development of tolerance and exacerbation of insomnia with rebound of REM sleep
upon withdrawal is also a major concern.82,83 Nonbenzodiazepine hypnotics have a better pharmacological profile with fewer and milder side effects
than the benzodiazepines, and are currently preferred for the treatment of sleep problems in affective disorders.61,84 Recently, ramelteon, a novel selective agonist of melatonin MT1 and MT2 receptors,
was approved for the treatment of primary insomnia. In comparison with the other hypnotics, it is
claimed that ramelteon has the advantage of no potential for abuse. However, experience with this
novel hypnotic agent in the treatment of insomnia
associated with depression is still limited.5
In cases resistant to treatment, the combination
of antidepressants and antipsychotics should be
considered. Adding an antipsychotic to the patients’
daily regimen (ie, olanzapine) might alleviate sleep
disturbances by increasing SWS.85
Conclusion
Sleep disturbances often occur in depressed patients,
and they may be used to identify newly presenting
or recurrent episodes of depression. Moreover, persistent insomnia is one of the most common residual symptoms in incompletely remitted depression.
Generally, complete relief of associated insomnia
should be pursued in patients under treatment with
antidepressants to avoid the recurrence of depressive symptomatology. Currently-available antidepressants have notable limitations, mainly relating
to their only moderate efficacy relative to placebo.
A combination of nonpharmacological interventions (sleep deprivation, light therapy, CBT, IPSRT)
together with pharmacotherapy could be used to
improve management of sleep disturbances associated with depression. A novel antidepressant compound, agomelatine, an agonist of melatonergic
MT1 and MT2 receptors as well as an antagonist of
5-HT2C receptors, has been recently introduced; it
shows a synchronizing effect on circadian rhythms
and demonstrates robust antidepressant efficacy,
while it produces a superior and earlier improvement in the sleep-wake cycle compared with other
antidepressant agents. This sleep-restorative antidepressant offers the possibility of an improved quality of remission, while minimizing relapses or recurrences of depressive episodes. Improvement of the sleep-wake cycle as a target for remission in depression – Soldatos and Theleritis
MEDICOGRAPHIA, VOL 31, No. 2, 2009 143
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
REFERENCES
1. Reynolds CF III, Kupfer DJ. Sleep research in affective illness:
state of the art circa 1987. Sleep. 1987;10:199-215.
2. Benca RM, Obermeyer WH, Thisted RA, Gillin JC. Sleep and
psychiatric disorders. A meta-analysis. Arch Gen Psychiatry.1992;
49:651-666.
3. Soldatos CR, Paparrigopoulos TJ. Sleep patterns in depression.
WPA Bulletin in Depression. 1996;3:4-5.
4. Argyropoulos SV, Wilson SJ. Sleep disturbances in depression
and the effects of antidepressants. Int Rev Psychiatry. 2005;17:
237-245.
5. Thase M. Depression and sleep: pathophysiology and treatment.
Dialogues Clin Neurosci. 2006;8:217-226.
6. Tsuno N, Besset A, Ritchie K. Sleep and depression. J Clin Psychiatry. 2005;66:1254-1269.
7. Lepine JP, Gastpar M, Mendlewicz J, Tylee A. Depression in the
community: the first pan-European Study DEPRES (Depression
Research in European Society). Int Clin Psychopharmacol.1997;
12:19-29.
8. Bixler EO, Kales A, Soldatos CR, Kales JD, Healey S. Prevalence
of sleep disorders in the Los Angeles Metropolitan area. Am J Psychiatry. 1979;135:1257-1262.
9. Mellinger GD, Balter MB, Uhlenhuth EH. Insomnia and its
treatment. Prevalence and correlates. Arch Gen Psychiatry.1985;
42:225-232.
10. Soldatos CR. Insomnia in relation to depression and anxiety:
epidemiologic considerations. J Psychosom Res. 1994;38:3-8.
11. Giles DE, Jarrett RB, Roffwarg HP, Rush AJ. Reduced rapid
eye movement latency. A predictor of recurrence in depression.
Neuropsychopharmacology. 1987;1:33-39.
12. Kennedy GJ, Kelman HR, Thomas C. Persistence and remission of depressive symptoms in late life. Am J Psychiatry.1991;
148:174-178.
13. Buysse DJ, Frank E, Lowe KK, Cherry CR, Kupfer DJ. Electroencephalographic sleep correlates of episode and vulnerability to
recurrence in depression. Biol Psychiatry. 1997;41:406-418.
14. Armitage R, Trivedi M, Hoffmann R, Rush AJ. Relationship
between objective and subjective sleep measures in depressed patients and healthy controls. Depress Anxiety. 1997;5:97-102.
15. Kupfer DJ, Frank E. The relationship of EEG sleep to vital depression. J Affect Disord. 1984;7:249-263.
16. Kupfer DJ, Foster FG, Coble P, McPartland RJ, Ulrich RF. The
application of EEG sleep for the differential diagnosis of affective
disorders. Am J Psychiatry. 1978;135:69-74.
17. Kupfer DJ, Ehlers CL. Two roads to rapid eye movement latency. Arch Gen Psychiatry. 1989;46:945-948.
18. Van Bemmel AL. Sleep, Depression and Antidepressants [thesis]. Maastricht, the Netherlands: Maastricht University; 1993.
19. McCarley RW. REM sleep and depression: common neurobiological control mechanisms. Am J Psychiatry.1982;139:565-570.
20. Janowsky DS, Davis JM, El-Yousef MK, et al. A cholinergicadrenergic hypothesis of mania and depression. Lancet. 1972;2:
632-635.
21. Vogel GW, Buffenstein A, Minter K, et al. Drug effects on REM
sleep and on endogenous depression. Neurosci Biobehav Rev.
1990;14:49-63.
22. Benca RM. Mood disorders. In: Kryger MH, Roth T, Dement
WC, eds. Principles and Practice of Sleep Medicine. 4th ed. Philadelphia, Pa: WB Saunders; 2005.
23. Steiger A. Sleep and the hypothalamic-pituitary-adrenal system. Sleep Med Rev. 2002;6:125-138.
24. Borbély AA, Wirz-Justice A. A two process model of sleep regulation. Hum Neurobiol. 1982;1:205-210.
25. Elsenga S, van den Hoofdakker RH. Clinical effects of several
sleep-wake manipulations on endogenous depression. Sleep Res.
1983;12:326.
26. Wirz-Justice A. Chronobiology and mood disorders. Dialogues
Clin Neurosci. 2003;5:315-325.
27. Schibler U, Ripperger J, Brown SA. Peripheral circadian oscillators in mammals: time and food. J Biol Rhythms. 2003;18:250260.
28. Roenneberg T, Wirz-Justice A, Merrow M. Life between clocks:
daily temporal patterns of human chronotypes. J Biol Rhythms.
2003;18:80-90.
29. Benedetti F, Serretti A, Colombo C, Lorenzi C, Tubazio V,
Smeraldi E. A glycogen synthase kinase 3-b promoter gene SNP
is associated with age at onset and response to total sleep deprivation in bipolar depression. Neurosci Lett. 2004;368:123-126.
30. Benedetti F, Serretti A, Colombo C, et al. Influence of CLOCK
gene polymorphism on circadian mood fluctuation and illness
recurrence in bipolar depression. Am J Med Genet. 2003;123B:
23-26.
31. Serretti A, Benedetti F, Mandelli L, et al. Genetic dissection of
psychopathological symptoms: insomnia in mood disorders and
144 MEDICOGRAPHIA, VOL 31, No. 2, 2009
CLOCK gene polymorphism. Am J Med Genet. 2003;121B:35-38.
32. Serretti A, Cusin C, Benedetti F, et al. Insomnia improvement
during antidepressant treatment and CLOCK gene polymorphism.
Am J Med Genet B Neuropsychiatr Genet. 2005;137:36-39.
33. Wirz-Justice A. Biological rhythm disturbances in mood disorders. Int Clin Psychopharmacol. 2006;21(suppl 1):S11–S15.
34. Mendlewicz J. Circadian rhythm disturbances in depression.
In: Mendlewicz J, ed. Circadian Rhythms and Depression: Current Understanding and New Therapeutic Perspectives. Paris,
France: Wolters Kluwer Health; 2008:21-35.
35. Wirz-Justice A, Terman M, Oren D, et al. Brightening depression. Science. 2004;303:467-468.
36. Wirz-Justice A, Benedetti F, Berger M, et al. Chronotherapeutics (light and wake therapy) in affective disorders. Psychol Med.
2005;35:939-944.
37. Wirz-Justice A, Van den Hoofdakker RH. Sleep deprivation
in depression: what do we know, where do we go? Biol Psychiatry. 1999;46:445-453.
38. Szuba MP, Baxter LR, Altshuler LL, et al. Lithium sustains
the acute antidepressant effects of sleep deprivation: preliminary
findings from a controlled study. Psychiatry Res.1994;51:283295.
39. Benedetti F, Colombo C, Barbini B, Campori E, Smeraldi E.
Ongoing lithium treatment prevents relapse after total sleep deprivation. J Clin Psychopharmacol. 1999;19:240-245.
40. Colombo C, Lucca A, Benedetti F, Barbini B, Campori E, Smeraldi E. Total sleep deprivation combined with lithium and light
therapy in the treatment of bipolar depression: replication of main
effects and interaction. Psychiatry Res. 2000;95:43-53.
41. Smeraldi E, Benedetti F, Barbini B, Campori E, Colombo C.
Sustained antidepressant effect of sleep deprivation combined
with pindolol in bipolar depression. A placebo-controlled trial.
Neuropsychopharmacology. 1999;20:380-385.
42. Lam RW, Levitt AJ. Canadian Consensus Guidelines for the
Treatment of Seasonal Affective Disorder. Vancouver, Canada:
Clinical and Academic Publishing; 1999.
43. Epperson CN, Terman M, Terman JS, et al. Randomized clinical trial of bright light therapy for antepartum depression: preliminary findings. J Clin Psychiatry. 2004;65:421-425.
44. Goel N, Terman M, Terman JS, Macchi MM, Stewart JW.
Controlled trial of bright light and negative air ions for chronic
depression. Psychol Med. 2005;35:945-955.
45. Benedetti F, Colombo C, Pontiggia A, Bernasconi A, Florita
M, Smeraldi E. Morning light treatment hastens the antidepressant effect of citalopram: a placebo-controlled trial. J Clin Psychiatry. 2003;64:648-653.
46. Martiny K. Adjunctive bright light in non-seasonal major depression. Acta Psychiatr Scand. 2004;(suppl 425):7-28.
47. Smith MT, Perlis ML, Park A, et al. Comparative meta-analysis of pharmacotherapy and behavior therapy for persistent insomnia. Am J Psychiatry. 2002;159:5-11.
48. Morin CM. Insomnia treatment: taking a broader perspective
on efficacy and cost-effectiveness issues. Sleep Med Rev. 2004;
8:3-6.
49. Feldman G. Cognitive and behavioral therapies for depression:
overview, new directions, and practical recommendations for dissemination. Psychiatr Clin North Am. 2007;30:39-50.
50. Smith MT, Huang MI, Manber R. Cognitive behavior therapy for chronic insomnia occurring within the context of medical
and psychiatric disorders. Clin Psychol Rev. 2005;25:559-592.
51. Frank E, Kupfer DJ, Thase ME, et al. Two-year outcomes for
interpersonal and social rhythm therapy in individuals with bipolar I disorder. Arch Gen Psychiatry. 2005;62:996-1004.
52. Germain A, Frank E. Chronobiology of the core symptoms of
depression. Medicographia. 2008;30:30-34.
53. Arendt J. Importance and relevance of melatonin to human
biological rhythms. J Neuroendocrinol. 2003;15:427-431.
54. Paparrigopoulos TJ, Theleritis CG, Soldatos CR. Sleep and antidepressant medication. WPA Bulletin in Depression. 2007;11:
7-11.
55. Lader M. Limitations of current medical treatments for depression: disturbed circadian rhythms as a possible therapeutic
target. European Neuropsychopharmacology. 2007;17:743-755.
56. Nelson JC, Portera L, Leon AC. Residual symptoms in depressed patients after treatment with fluoxetine or reboxetine.
J Clin Psychiatry. 2005;66:1409-1414.
57. Paykel ES. Remission and residual symptomatology in major
depression. Psychopathology. 1998;31:5-14.
58. Thase ME, Simons AD, McGeary J, et al. Relapse after cognitive behavior therapy of depression: potential implications for
longer courses of treatment? Am J Psychiatry. 1992;149:10461052.
59. Nutt D, Wilson S, Paterson L. Sleep disorders as core symp-
Improvement of the sleep-wake cycle as a target for remission in depression – Soldatos and Theleritis
TRUE REMISSION
toms of depression. Dialogues Clin Neurosci. 2008;10:329-338.
60. Mayers AG, Baldwin DS. Antidepressants and their effect on
sleep. Hum Psychopharmacol Clin Exp. 2005;20:533-559.
61. Riemann D, Voderholzer U, Cohrs S, et al. Trimipramine in
primary insomnia: results of a polysomnographic double-blind
controlled study. Pharmacopsychiatry. 2002;35:165-174.
62. Volkers AC, Tulen JHM, van den BroekWW, Bruijn JA, Passchier J, Pepplinkhuizen L. 24-Hour motor activity after treatment
with imipramine or fluvoxamine in major depressive disorder.
Eur Neuropsychopharmacol. 2002;12:273-278.
63. Shipley JE, Kupfer DJ, Griffin SJ, et al. Comparison of effects
of desipramine and amitriptyline on EEG sleep of depressed patients. Psychopharmacology. 1985;85:14-22.
64. McCarley RW. REM sleep and depression: common neurobiological control mechanisms. Am J Psychiatry. 1982;139:565570.
65. Kupfer DJ, Spiker DG, Coble PA, Neil JF, Ulrich R, Shaw DH.
Sleep and treatment prediction in endogenous depression. Am J
Psychiatry. 1981;138:429-434.
66. Wilson SJ, Argyropoulos SV. Antidepressants and sleep: a qualitative review of the literature. Drugs. 2005;65:927-947.
67. Gursky JT, Krahn LE. The effects of antidepressants on sleep:
a review. Harv Rev Psychiatry. 2000;8:298-306.
68. Qureshi A, Lee-Chiong JR. Medications and their effects on
sleep. Med Clin North Am. 2004;88:751-766.
69. Saletu B, Frey R, Krupka M, Anderer P, Grunberger J, See WR.
Sleep laboratory studies on the single-dose effects of serotonin reuptake inhibitors paroxetine and fluoxetine on human sleep and
awakening qualities. Sleep. 1991;14:439-447.
70. Salin-Pasqual RJ, Galizia-Polo L, Drucker-Colin A. Sleep
changes after 4 consecutive days of venlafaxine administration
in normal volunteers. J Clin Psychiatry. 1997;58:348-350.
71. Luthringer R, Toussaint M, Schaltenbrand M, et al. A double
blind placebo controlled evaluation of the effects of orally administered venlafaxine on sleep in patients with major depression.
Psychopharmacol Bull. 1996;32:637-646.
72. Chalon S, Pereira A, Lainey E, et al. Comparative effects of
duloxetine and desipramine on sleep EEG in healthy subjects.
Psychopharmacology. 2005;177:357-365.
73. Sartorius N, Baghai TC, Baldwin DS, et al. Antidepressant
medications and other treatments of depressive disorders: a CINP
Task Force report based on a review of evidence. Int J Neuropsy-
IN
DEPRESSION: T
HE
ULTIMATE GOAL
chopharmacol. 2007;10(suppl 1):S1-S207.
74. Löo H, D’haenen H, Hale A. Determination of the dose of
agomelatine, a melatoninergic agonist and selective 5-HT2c antagonist, in the treatment of major depressive disorder: a placebo-controlled dose range study. Int J Neuropsychopharmacol.
2002;17:239-247.
75. Kennedy SH, Emsley R. Placebo-controlled trial of agomelatine in the treatment of major depressive disorder. Eur Neuropsychopharmacol. 2006;16:93-100.
76. Olié JP, Kasper S. Efficacy of agomelatine, a MT1/MT2 receptor agonist with 5-HT2C antagonistic properties, in major depressive disorder. Int J Neuropsychopharmacol. 2007;10:661-673.
77. Cajochen C, Kräuchi K, Möri D, Graw P, Wirz-Justice A. Melatonin and S-20098 increase REM sleep and wake-up propensity
without modifying NREM sleep homeostasis. Am J Physiol Regul Integr Comp Physiol. 1997;272:R1189–R1196.
78. Kräuchi K, Cajochen C, Möri D, Graw P, Wirz-Justice A. Early evening melatonin and S-20098 advance circadian phase and
nocturnal regulation of core body temperature. Am J Physiol
Regul Integr Comp Physiol. 1997;272:R1178–R1188.
79. Millan MJ, Gobert A, Lejeune F, et al. The novel melatonin
agonist agomelatine (S20098) is an antagonist at 5-HT2C receptors, blockade of which enhances the activity of frontocortical
dopaminergic and adrenergic pathways. J Pharmacol Exp Ther.
2003;306:954-964.
80. Birkenhager TK, Moleman P, Nolen WA. Benzodiazepines for
depression? A review of the literature. Int Clin Psychopharmacol.
1995;10:181-195.
81. McCall WV. A psychiatric perspective on insomnia. J Clin Psychiatry. 2001;62(suppl 10):27-32.
82. Schweitzer PK. Drugs that disturb sleep and wakefulness.
In: Kryger MH, Roth T, Dement WC, eds. Principles and Practice
of Sleep Medicine. 3rd ed. Philadelphia, Pa: WB Saunders; 2000:
441-461.
83. Soldatos CR, Dikeos DG, Whitehead A. Tolerance and rebound
with rapidly eliminating hypnotics: a meta-analysis of sleep laboratory studies. Int Clin Psychopharmacol. 1999;14:287-303.
84. Obermeyer W, Benca R. Effects of drugs on sleep. Neurol Clin.
1996;14:827-840.
85. Sharpley AL, Attenburrow MEJ, Hafizi S, Cowen PJ. Olanzapine increases slow wave sleep and sleep continuity in SSRI-resistant depressed patients. J Clin Psychiatry. 2005;66:450-454.
L’AMÉLIORATION
DU CYCLE VEILLE-SOMMEIL COMME CIBLE
DE LA RÉMISSION DANS LA DÉPRESSION
L
es troubles du sommeil sont associés depuis longtemps à la dépression ;
souvent utilisés pour identifier les patients débutant une dépression, ils
pourraient appartenir à une perturbation plus générale des rythmes
biologiques. De plus, l’insomnie persistante est un symptôme résiduel fréquent
d’une dépression incomplètement guérie. Afin d’éviter la récidive des symptômes dépressifs, il est de règle de poursuivre le traitement de l’insomnie chez
les patients traités par antidépresseurs. L’association de traitements médicamenteux et non médicamenteux ciblant le cycle veille-sommeil (privation de
sommeil, luminothérapie) peut contribuer à la disparition des symptômes dépressifs et des troubles du sommeil associés. L’agomélatine, une nouvelle molécule agoniste des récepteurs mélatoninergiques MT1 et MT2 et antagoniste des
récepteurs 5-HT2C , possède un effet unique de synchronisation des rythmes
circadiens et une bonne efficacité antidépressive.
Improvement of the sleep-wake cycle as a target for remission in depression – Soldatos and Theleritis
MEDICOGRAPHIA, VOL 31, No. 2, 2009 145
TRUE REMISSION
IN
DEPRESSION: THE ULTIMATE GOAL
Remission: concept and reality
R
Sidney H. KENNEDY, MD, FRCPC
Sakina RIZVI, HBSc
Department of Psychiatry
University Health Network
University of Toronto
Toronto, CANADA
Sexual functioning
and quality of life in
patients in remission
from depression
by S. H. Kennedy and
S. Rizvi, Canada
he adoption of remission as a clinical outcome in the treatment of major
depressive disorder (MDD) has served to emphasize the important adverse
consequences of residual symptoms following antidepressant treatment.
These limitations are further underscored by differences between clinician and
patient perceptions of remission, highlighting the need to extend outcome measures to include a broader evaluation of quality of life in patients who have
received treatment for MDD. Sexual dysfunction is frequently reported as a
symptom of MDD and more frequently occurs as a result of many current antidepressant treatments. The consequences of sexual dysfunction in many depressed patients include impairment of quality of life as well as treatment nonadherence and ultimately drug discontinuation. Many antidotes to alleviate
antidepressant-induced sexual dysfunction have been advocated, but few have
robust, randomized controlled trial evidence to support their recommendation.
Where possible, monotherapy with an antidepressant that respects sexual function is a preferred strategy.
T
Medicographia. 2009;31:146-151.
(see French abstract on page 151)
Keywords: sexual dysfunction; remission; quality of life; antidepressant;
nonadherence; discontinuation
www.medicographia.com
Address for correspondence: Professor Sidney Kennedy, Psychiatrist-in-Chief,
University Health Network, University of Toronto, Toronto, Canada
(e-mail: [email protected])
146 MEDICOGRAPHIA, VOL 31, No. 2, 2009
emission in major depressive disorder (MDD)
typically refers to symptom remission; that
is, the near absence of all pre-existing symptoms endorsed on an established rating scale for
depressive symptoms.1 Virtually all studies on the
treatment of a major depressive episode utilize either the 17-item Hamilton Rating Scale for Depression (HAM-D17)2 or the Montgomery–Åsberg Depression Rating Scale (MADRS)3 to assess remission.
For the HAM-D17, a score of 7 or less to connote remission has achieved the greatest acceptance.4 On
MADRS, scores of 10 or less, or in some instances 12
or less, have been advocated to reflect remission.5,6
Evidence shows that there is variation among
remission rates for different antidepressants and
in different patient populations. Higher remission
rates have been reported with venlafaxine (40%45%) compared with selective serotonin reuptake
inhibitors (SSRIs; 35%-40%) in some,7,8 but not all,
meta-analyses of randomized controlled trials.9,10
Patients in effectiveness trials typically achieve even
lower rates of remission. In the Sequenced Treatment Alternatives to Relieve Depression (STAR*D)
study,11 only 28% of patients who received citalopram (SSRI) for up to 14 weeks met remission criteria.
Failure to achieve remission is associated with
many adverse consequences, including high rates
of recurrence, decreased work productivity, greater
medical comorbidity, and an overall decrease in
quality of life.4,12 Even patients treated to remission report a substantial burden of subthreshold
or threshold symptoms: insomnia (44%), fatigue
(38%), and anhedonia (27%) were the most prevalent residual symptoms in fluoxetine-treated remitters,13 while anxiety and loss of libido were also
prevalent in other patient populations following
remission.14,15 Although clinically meaningful, a limitation in the concept of remission is that it may not
fully reflect the patient’s perspective regarding the
balance between symptom change, side effect burden, and functional outcome. Patients reported
“presence of positive mental health,” “return to
one’s usual normal self,” and “return to usual level
of functioning” as the three most important items
in determining remission.16
SELECTED
ABBREVIATIONS AND ACRONYMS
DSM-IV TR Diagnostic and Statistical Manual of
Mental Disorders - Fourth Edition
Text Revision
HAM-D17
17-item Hamilton Rating Scale for
Depression
ICD-10
International Classification of Diseases and Related Health Problems 10th Edition
MADRS
Montgomery–Åsberg Depression
Rating Scale
MDD
Major Depressive Disorder
SSRI
selective serotonin reuptake inhibitor
STAR*D
Sequenced Treatment Alternatives to
Relieve Depression (study)
Sexual functioning and quality of life in patients in remission from depression – Kennedy and Rizvi
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
Classification and measurement
Symptoms
Sexual dysfunction
Weight gain
Fatigue
Anticholinergic effects
Agitation
Insomnia
Gastrointestinal upset
Headache
N
%
231
201
107
82
73
48
28
2
20
18
9
7
6
4
3
<1
Table I. Symptoms influencing psychiatrists’ antidepressant choice for outpatients.
Modified from reference 21: Zimmerman M, Posternak M, Friedman
M, et al. Which factors influence psychiatrists’ selection of antidepressants? Am J Psychiatry. 2004;161:1285-1289. Copyright ©
2004, American Psychiatric Association.
Quality of life as an expanded concept
By assessing a patient’s overall sense of satisfaction
and personal wellbeing, quality of life measures reflect outcomes beyond symptom improvement. Patients with MDD may experience limitations in the
areas of physical health, work efficiency, and social
interactions. Measures have been developed for use
across a wide range of disorders and populations
(eg, Euro Quality of Life-5D, Short-Form 36-Item
Questionnaire), across different psychiatric populations (eg, Social Adjustment Scale, Quality of Life
Enjoyment and Satisfaction Questionnaire), or specifically, in depressed patients (Social Adaptation
and Self-Evaluation Scale).17 These scales have been
valuable in demonstrating that psychosocial impairment persists in MDD even beyond remission,18,19
indicating that symptom improvement and quality of life do not necessarily improve at the same
rate. When MDD patients were evaluated periodically over 5 years of treatment, marital and sexual
satisfaction ratings were persistently lower compared with controls.20 Achieving symptomatic remission has to be partnered with minimal side effect burden to attain the best treatment outcome.
This was recognized as an important issue by psychiatrists, who ranked sexual dysfunction as the
highest consideration in prescribing an antidepressant, followed by weight gain and fatigue (Table I).21
Sexual dysfunction as a component
of remission and quality of life
In addition to sleep and alertness22 and neurocognitive symptoms,23 healthy sexual function is an
important component of good quality of life, yet
sexual dysfunction is a frequent consequence of antidepressant therapy,24-26 and is unacceptable to many
patients. In a large study involving over 6000 patients who received antidepressant therapy, 85%
rated sexual functioning as “extremely important,”
“very important,” or “important,” while only 3% felt
it was “not important.”24 Unfortunately, sexual dysfunction is a relatively neglected aspect of both remission and quality of life, despite its simple measurement, high prevalence, and negative impact on
treatment adherence.
The International Classification of Diseases and
Related Health Problems–10th Edition (ICD-10)27
lists 10 forms of sexual dysfunction (Table II), which
can be associated with physical or psychiatric conditions, medication treatments, or psychosocial factors. Sexual dysfunction in MDD typically involves
problems with desire, arousal or orgasmic/ejaculatory function, and may occur across several of these
areas. While the Diagnostic and Statistical Manual of Mental Disorders–Fourth Edition Text Revision (DSM IV-TR) does not explicitly include loss
of sexual interest or desire as a core item, clinicians
consider libido as a component of “diminished interest or pleasure,” and a single item on HAM-D17
captures desire or interest in sex as a component of
overall symptom severity. However, this scale does
not assess the broader impact of sexual dysfunction
on quality of life.
The issue of measurement across the spectrum
of sexual dysfunction is of particular importance
when considering the adverse impact that SSRIs
and other antidepressants can have on diverse ar Lack or loss of sexual desire
Sexual aversion and lack of sexual enjoyment
Failure of genital response
Orgasmic dysfunction
Premature ejaculation
Nonorganic vaginismus
Nonorganic dyspareunia
Excessive sexual drive
Other sexual dysfunction, not caused by organic
disorder of disease
Unspecified sexual dysfunction, not caused by
organic disorder or disease
Table II. Classification of sexual dysfunction according
to the International Classification of Mental and Behavioural Disorders–10th edition. Based on data from
reference 27.
eas of sexual function.28,29 Several easy to use sexual
function scales are available (Arizona Sexual Experience Scale, Changes in Sexual Functioning Questionnaire, Psychotropic-Related Sexual Dysfunction
Questionnaire, and the Sex Effects Scale).25 The importance of administering direct assessment scales
is underscored by the findings that symptom reports increase twofold (69%) with direct questioning compared with reliance on spontaneous self-report (35%).30
Prevalence and risk factors
The prevalence of sexual dysfunction in the community is estimated to be approximately 20%30%31,32: a considerably higher prevalence rate of
50% or more has been reported in untreated depressed patients, and even higher rates are reported during antidepressant treatment.30 Risk factors
for sexual dysfunction among psychiatric inpatients
include: (i) history of physical, emotional, and sexual abuse; (ii) self-harm and depression; (iii) poor
self-rated health and cardiopulmonary conditions;
and (iv) use of antidepressants.33
Sexual functioning and quality of life in patients in remission from depression – Kennedy and Rizvi
MEDICOGRAPHIA, VOL 31, No. 2, 2009 147
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
Adherence and discontinuation
Treatment-emergent sexual dysfunction can cause
significant distress to depressed patients, which adversely affects quality of life34 and leads to antidepressant nonadherence.35,36 Sexual dysfunction was
the most bothersome side effect in a survey of more
than 400 patients treated with SSRIs for approximately 3 months.37 Beside being a troublesome adverse event, sexual dysfunction is also one of the
most common side effects leading to treatment
discontinuation during short-term and long-term
care.38,39 Further supporting this finding, Rosenberg and colleagues40 reported that 42% of men and
15% of women discontinue antidepressant treatment over perceived beliefs about sexual side effects.
Managing drug-induced
sexual dysfunction in remitted patients
In order to reduce treatment nonadherence, several strategies can be used to mitigate treatmentemergent sexual dysfunction, including augmentation with an antidote, switching medications, or
preferably prescribing an antidepressant with fewer sexual side effects in the first instance. Although
primary prevention is the optimal solution, this is
not always possible in the treatment of MDD. Several antidepressants are associated with treatmentemergent sexual dysfunction in less than 10% of patients, while others result in significant impairment
in 30% or more patients (Table III).41 These differences have been linked to putative neurotransmitter and neuropeptide abnormalities associated with
sexual dysfunction, and the beneficial or adverse
effects of various antidepressants on these targets.
Four mechanisms associated with drug-induced
sexual dysfunction involve: (i) decreased dopaminergic activity; (ii) agonism of 5HT2 and 5HT3 receptors; (iii) blockade of adrenergic and cholinergic
receptors; and (iv) inhibition of nitric oxide synthase. Various antidotes targeting one or more of
these mechanisms have been advocated to mitigate
these adverse effects.
Antidotes to mitigate sexual dysfunction
Enhancement of dopaminergic activity
Bupropion, psychostimulants, and amantadine have
all been evaluated as treatments to relieve sexual
dysfunction. In two out of three bupropion add-on
studies, a dose of 150 mg versus placebo failed to improve sexual dysfunction associated with SSRIs,42,43
although augmentation with bupropion SR 150 mg
twice daily was significantly better than placebo in
improving desire and frequency of sexual activity
in remitted depressed patients on SSRI therapies.44
It was also observed in an open-label augmentation
study that bupropion not only improved sexual dysfunction in venlafaxine-treated patients, but also increased blood levels of venlafaxine approximately
threefold.45
Stimulants such as methylphenidate have also
been advocated to treat sexual dysfunction on the
basis of their prodopaminergic effects. Although
augmentation of antidepressant monotherapy with
sustained release methylphenidate did not signifi148 MEDICOGRAPHIA, VOL 31, No. 2, 2009
cantly enhance depression outcomes, there was a
significant improvement in sexual function, as well
as apathy and fatigue, compared with placebo.46 In
clinical practice, other stimulants are used to enhance sexual function (dextroamphetamine, modafanil), but there are no randomized-controlled trials
to support these recommendations. On a cautionary note, various dopamine agonists used to treat
Parkinson’s disease (pramipexole, ropinirole, and
pergolide) have been associated with pathological
hypersexuality.47
Antagonism of 5HT2 and 5HT3 receptors or
agonism of 5HT1A receptors
Several antidepressants are known to display antagonist effects at 5HT2 or 5HT3 receptors. In the
case of mirtazapine, there was found to be no advantage over placebo when the drug was added to
fluoxetine in depressed women experiencing druginduced sexual dysfunction.48 There is preliminary
evidence that mirtazapine added to duloxetine improves duloxetine-induced sexual dysfunction,49 and
<10%
Agomelatine
Bupropion
Milnacipran
Mirtazapine
Moclobemide
Reboxetine
10%-30%
>30%
Citalopram
Duloxetine
Escitalopram
Venlafaxine
Fluoxetine
Fluvoxamine
Paroxetine
Sertraline
Table III. Frequency of sexual dysfunction during antidepressant treatment with different antidepressant
medications. Based on data from reference 41.
anecdotal support for cyproheptadine, a 5HT2 antagonist, particularly for anorgasmia,50,51 although
granisetron, a 5HT3 antagonist, did not offer any advantage over placebo.52
The potentially beneficial antagonism of 5HT2A
and 5HT2C receptors by atypical antipsychotics is
countered by other mechanisms likely to increase
serotonergic output and exacerbate sexual dysfunction. Despite the increasing use of “atypicals” such
as risperidone, olanzapine, aripiprazole, quetiapine,
and ziprasidone to treat patients with mood disorders, only minimal attention has been paid to sexual function. There was found to be no significant
advantage of olanzapine versus placebo in reversing
antidepressant-induced side effects,48 and contradictory findings were reported in two small openlabel studies using risperidone: a reduction in sexual function in one study and potentially beneficial
effects in the other.53,54 The only systematic comparison of sexual dysfunction across atypical antipsychotics was performed in a large cross-sectional
study involving more than 600 schizophrenia patients. Sexual dysfunction was reported in 43% of
patients receiving risperidone, 35% on olanzapine,
and 18% on quetiapine.55 Differences in diagnosis,
underlying neurotransmitter function, dosing of
atypical antipsychotics, and baseline sexual function limit the generalizability of these findings to
the MDD population. Several agents with 5HT1A agonist properties have been evaluated as antidotes
Sexual functioning and quality of life in patients in remission from depression – Kennedy and Rizvi
TRUE REMISSION
for sexual dysfunction. The anxiolytic, buspirone,
has been shown to improve SSRI-induced sexual
dysfunction, with more pronounced effects in women than men.56 Though flibanserin failed to demonstrate antidepressant efficacy, this 5HT1A agonist
and 5HT2A antagonist with minimal DA4 agonist
effects57 has been evaluated for the treatment of Hypoactive Sexual Desire Disorder in women, with
positive results.58 VML-670 is a novel 5HT1A agonist
with weak 5HT1D agonist properties. It was not superior to placebo in overall improvement of sexual
dysfunction in previously depressed male and female patients.59
Blockade of adrenergic and cholinergic receptors
Yohimbine is an α-adrenergic agonist with favorable anecdotal evidence about its role in treating
sexual dysfunction, but negative results under placebo-controlled conditions.48 Bethanecol has mixed
central and peripheral cholinergic and adrenergic
effects. It was effective in reversing clomipramineinduced ejaculatory delay under placebo-controlled
conditions.60
Inhibition of nitric oxide synthase
The largest trials and most robust evidence for reversal of SSRI-induced sexual dysfunction involve
treatment with sildenafil, the prototypic phosphodiesterase inhibitor. Sildenafil in doses of 25-100 mg
has been shown to significantly enhance sexual function in both men61,62 and women63 treated to remission with SSRIs. Preliminary evidence also supports
the role of tadalafil in reversing SSRI-induced sexual dysfunction in both men64 and women.65
Comparison of sexual dysfunction across
antidepressant monotherapies
Sexual dysfunction is a significant problem with
first generation monoamine oxidase inhibitors and
tricyclic antidepressants, although other adverse effects have been of much greater clinical concern.
The consensus from a series of well-designed
comparative studies is that up to 60% of patients
receiving SSRIs report some form of treatmentemergent sexual dysfunction.24,28,66-71 All of the SSRIs
have been associated with delayed or absent orgasm/
ejaculation, and in some instances, a reduction in
libido and arousal.28,72,73
Similar rates of sexual dysfunction have been reported with serotonin norepinephrine reuptake
inhibitors,68,74 although some evidence supports a
lower rate with duloxetine, desvenlafaxine, and milnacipran, potentially related to greater noradrenergic blockade.75-77 Reboxetine is the only second
generation norepinephrine receptor inhibitor, and
is not widely available as an antidepressant. Nevertheless, it is associated with a relatively favorable
sexual function profile compared with SSRIs.78
Among antidepressant monotherapies with the
least likelihood of inducing sexual dysfunction, other variables beyond efficacy need to be considered.
In the case of mirtazapine, switching to this from
REFERENCES
1. Frank E, Prien RF, Jarrett RB, et al. Conceptualization and rationale for consensus definitions of terms in major depressive
disorder. Remission, recovery, relapse, and recurrence. Arch Gen
Psychiatry. 1991;48:851-855.
IN
DEPRESSION: T
HE
ULTIMATE GOAL
an SSRI resulted in lower rates of sexual dysfunction,79 but other adverse effects including weight
gain and daytime sedation resulted in high rates of
discontinuation.48 Nefazodone, a 5HT2 receptor antagonist and norepinephrine reuptake inhibitor,
was relatively sparing of sexual function,80 but is
now largely unavailable because of hepatotoxicity
concerns.81 Bupropion, with its synergistic effects
on dopamine and norepinephrine systems, results
in less sexual dysfunction than SSRIs 5,82 or venlafaxine.74 Similarly, moclobemide, a reversible inhibitor
of monoamine oxidase A, has a favorable impact on
sexual dysfunction, although it is not generally considered to be a first-line antidepressant.28,83,84
Agomelatine, a novel antidepressant with direct
agonist effects on the melatonergic MT1 and MT2
receptors, as well as 5HT2C antagonist properties,85
has favorable effects on sexual function. Both mechanisms may synergistically account for its lack of
sexual dysfunction, since agomelatine exerts putative antagonistic effects on 5HT2 receptors, which
are in turn involved in sexual behaviour.86 In addition to antidepressant efficacy,87-89 agomelatine has
been evaluated in depressed patients and healthy
volunteers with sexual function as a primary outcome measure.5,90 Among sexually active remitted
patients, there was significantly less treatmentemergent sexual dysfunction in patients receiving
agomelatine compared with venlafaxine, specifically in the domains of sexual drive/desire in men and
orgasm in women. This was illustrated by significantly greater deteriorations on the Sex Effects scale
with venlafaxine for sexual drive/desire in men and
orgasm in women (an approximately fivefold difference compared with agomelatine for both).5 In other agomelatine trials in which the Arizona Sexual
Experience Scale was used, the rate of sexual dysfunction with agomelatine was comparable to or
lower than that reported in the placebo group. These
results are supported by a study carried out in
healthy male volunteers who were randomized to
receive agomelatine, paroxetine, or placebo for 8
weeks. The percentage of volunteers who experienced severe or moderate sexual dysfunction was
under 5% for agomelatine, 62% for paroxetine, and
0% for placebo.90
Conclusion
The narrow clinical concept of remission, while
valuable in a number of ways, fails to capture the
broader domains associated with good quality of
life. Healthy sexual function is an important outcome for patients with MDD, yet the majority of
currently prescribed antidepressants induce or exacerbate sexual dysfunction. While there are various
pharmacological approaches to mitigate drug-induced sexual dysfunction, prescribing a first-line
antidepressant treatment that respects sexual function is a preferred approach. 2. Hamilton M. A rating scale for depression. J Neurol Neurosurg
Psychiatry. 1960;23:56-62.
3. Montgomery SA, Åsberg M. A new depression scale designed to
be sensitive to change. Brit J Psych. 1979;134:382-389.
Sexual functioning and quality of life in patients in remission from depression – Kennedy and Rizvi
MEDICOGRAPHIA, VOL 31, No. 2, 2009 149
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
4. Thase ME. Introduction: defining remission in patients treated with antidepressants. J Clin Psychiatry. 1999;60(suppl 22):3-6.
5. Kennedy SH, Rizvi S, Fulton K, et al. A double-blind comparison of sexual functioning, antidepressant efficacy, and tolerability between agomelatine and venlafaxine XR. J Clin Psychopharmacol. 2008;28:329-333.
6. Thase ME, Entsuah AR, Rudolph RL. Remission rates during
treatment with venlafaxine or selective serotonin reuptake inhibitors. Br J Psychiatry. 2001;178:234-241.
7. Möller HJ. Outcomes in major depressive disorder: the evolving concept of remission and its implications for treatment. World
J Biol Psychiatry. 2008;9:102-114.
8. Nemeroff CB, Entsuah R, Benattia I, et al. Comprehensive analysis of remission (COMPARE) with venlafaxine versus SSRIs. Biol
Psychiatry. 2008;63:424-434.
9. Kennedy SH, Andersen HF, Thase ME. Escitalopram in the
treatment of major depressive disorder: a meta-analysis. Clin Med
Res Opin. In Press.
10. Weinmann S, Becker T, Koesters M. Re-evaluation of the efficacy and tolerability of venlafaxine vs SSRIs: meta-analysis. Psychopharmacol. 2008;196:511-520.
11. Trivedi MH, Rush AJ, Wisniewski SR, et al. Evaluation of outcomes with citalopram for depression using measurement-based
care in STAR*D: implications for clinical practice. Am J Psychiatry. 2006;163:28-40.
12. McIntyre RS, O'Donovan C. The human cost of not achieving
full remission in depression. Can J Psychiatry. 2004;49(3 suppl
1):10S-16S.
13. Nierenberg AA, Keefe BR, Leslie VC, et al. Residual symptoms
in depressed patients who respond acutely to fluoxetine. J Clin
Psychiatry. 1999;60:221-225.
14. Opdyke KS, Reynolds CF III, Frank E, et al. Effect of continuation treatment on residual symptoms in late-life depression:
how well is “well”? Depress Anxiety. 1996;4:312-319.
15. Paykel ES, Ramana R, Cooper Z, et al. Residual symptoms after partial remission: an important outcome in depression. Psychol Med. 1995;25:1171-1180.
16. Zimmerman M, McGlinchey JB, Posternak MA, et al. How
should remission from depression be defined? The depressed patient’s perspective. Am J Psychiatry. 2006;163:148-150.
17. Kennedy SH, Eisfeld BS, Cooke RG. Quality of life: an important dimension in assessing the treatment of depression? J Psychiatry Neurosci. 2001;26(suppl)S23-S28.
18. Agosti V, Stewart JW. Social functioning and residual symptomatology among outpatients who responded to treatment and
recovered from major depression. J Affect Disord. 1998;47:207210.
19. Keller MB, Lavori PW, Friedman B, et al. The longitudinal
follow-up evaluation: a comprehensive method for assessing outcome in prospective longitudinal studies. Arch Gen Psychiatry.
1987;44:540-548.
20. Coryell W, Scheftner W, Keller M, et al. The enduring psychosocial consequences of mania and depression. Am J Psychiatry.
1993;150:720-727.
21. Zimmerman M, Posternak M, Friedman M, et al. Which factors influence psychiatrists' selection of antidepressants? Am J
Psychiatry. 2004;161:1285-1289.
22. Soldatos CR, Theleritis CG. Sleep-wake cycle quality as a target for remission in depression. Medicographia. 2009;31:140145.
23. Price J, Goodwin GM. Emotional blunting or reduced reactivity following remission of major depression. Medicographia.
2009;31:152-156.
24. Clayton A, Pradko J, Croft H, et al. Prevalence of sexual dysfunction among newer antidepressants. J Clin Psychiatry. 2002;
63:357-366.
25. Kennedy SH, Rizvi S. Sexual dysfunction, depression and the
impact of antidepressants. J Clin Psychopharmacol. In press.
26. Montgomery SA, Baldwin DS, Riley A. Antidepressant medications: a review of the evidence for drug-induced sexual dysfunction. J Affect Disord. 2002;69:119-140.
27. World Health Organization. Clinical descriptions and diagnostic guidelines. The ICD-10 Classification of Mental and Behavioral Disorders. Geneva, Switzerland: World Health Organization; 1992.
28. Montejo A, Llorca G, Izquierdo J, et al. Incidence of sexual dysfunction associated with antidepressant agents: a prospective
multicenter study of 1022 outpatients. J Clin Psychiatry. 2001;62:
10-21.
29. Williams VS, Baldwin DS, Hogue SL, et al. Estimating the
prevalence and impact of antidepressant-induced sexual dysfunction in 2 European countries: a cross-sectional patient survey. J Clin Psychiatry. 2006;67:204-210.
30. Bonierbale M, Lançon C, Tignol J. The ELIXIR study: evalu-
150 MEDICOGRAPHIA, VOL 31, No. 2, 2009
ation of sexual dysfunction in 4557 depressed patients in France.
Curr Med Res Opin. 2003;19:114-124.
31. Angst J. Sexual problems in healthy and depressed persons.
Int Clin Pharmacol. 1998;13:S1-S4.
32. Laumann EO, Paik A, Rosen RC. Sexual dysfunction in the
United States: prevalence and predictors. JAMA. 1999;281:537544.
33. Perlman CM, Martin L, Hirdes JP, Curtin-Telegdi N, Perez E.
Prevalence and predictors of sexual dysfunction in psychiatric
inpatients. Psychosomatics. 2007;48:309-318.
34. Derogatis L. Sexual function and quality of life: endpoints and
outcomes. J Gend Specif Med. 2001;4:35-42.
35. Gregorian R, Golden K, Bahce A, et al. Antidepressant-induced
sexual dysfunction. Ann Pharmacother. 2002;36:1577-1589.
36. Zajecka J. Clinical issues in long-term treatment with antidepressants. J Clin Psychiatry. 2000;61:20-25.
37. Hu XH, Bull SA, Hunkeler EM. Incidence and duration of
side effects and those rated as bothersome with selective serotonin
reuptake inhibitor treatment for depression: patient report versus physician estimate. J Clin Psychiatry. 2004;65:959-965.
38. Bull SA, Hunkeler EM, Lee JY. Discontinuing or switching
serotonin reuptake inhibitors. Ann Pharmacother. 2002;36:578584.
39. van Geffen EC, van der Wal SW, van Hulten R, et al. Evaluation of patients’ experiences with antidepressants reported by
means of a medicine reporting system. Eur J Clin Pharmacol.
2007;63:1193-1199.
40. Rosenberg KP, Bleiberg KL, Koscis J, Gross C. A survey of
sexual side effects among severely mentally ill patients taking psychotropic medications: impact on compliance. J Sex Marital Ther.
2003;29:289-296.
41. Kennedy SH, Lam RW, Nutt J, Thase ME. Treating Depression
Effectively. 2nd ed. Oxfordshire, UK: Informa Healthcare; 2007.
42. Masand PS, Ashton AK, Gupta S, Frank B. Sustained-release
bupropion for selective serotonin reuptake inhibitor-induced sexual dysfunction: a randomized, double-blind, placebo-controlled,
parallel-group study. Am J Psychiatry. 2001;158:805-807.
43. DeBattista C, Solvason B, Poirier J, Kendrick E, Loraas E. A
placebo-controlled, randomized, double-blind study of adjunctive bupropion sustained release in the treatment of SSRI-induced
sexual dysfunction. J Clin Psychiatry. 2005;66:844-848.
44. Clayton AH, Warnock JK, Kornstein SG, et al. A placebo-controlled trial of bupropion SR as an antidote for selective serotonin
reuptake inhibitor-induced sexual dysfunction. J Clin Psychiatry.
2004;65:62-67.
45. Kennedy SH, McCann SM, Masellis M, et al. Combining bupropion SR with venlafaxine, paroxetine, or fluoxetine: a preliminary report on pharmacokinetic, therapeutic, and sexual dysfunction effects. J Clin Psychiatry. 2002;63:181-186.
46. Ravindran AV, Kennedy SH, O’Donovan MC, et al. Osmoticrelease oral system methylphenidate augmentation of antidepressant monotherapy in major depressive disorder: results of a
double-blind, randomized, placebo-controlled trial. J Clin Psychiatry. 2008;69:87-94.
47. Klos KJ, Bower JH, Josephs KA, Matsumoto JY, Ahlskog JE.
Pathological hypersexuality predominantly linked to adjuvant
dopamine agonist therapy in Parkinson’s disease and multiple
system atrophy. Parkinsonism Relat Disord. 2005;11:381-386.
48. Michelson D, Kociban K, Tamura R, Morrison MF. Mirtazapine, yohimbine or olanzapine augmentation therapy for serotonin
reuptake-associated female sexual dysfunction: a randomized,
placebo controlled trial. J Psychiatr Res. 2002;36:147-152.
49. Ravindran LN, Eisfeld BS, Kennedy SH. Combining mirtazapine and duloxetine in treatment-resistant depression improves
outcomes and sexual function. J Clin Psychopharmacol. 2008;
28:107-108.
50. Aizenberg D, Zemishlany Z, Weizman A. Cyprtoheptadine
treatment of sexual dysfunction induced serotonin reuptake inhibitors. Clin Neuropsychopharmacol. 1995;4:320-324.
51. Lauerma H. Successful treatment of citalopram-induced
anorgasmia by cyproheptadine. Acta Psychiatr Scand.1996;93:
69-70.
52. Nelson EB, Shah VN, Welge JA, Keck PE Jr. A placebo-controlled, crossover trial of granisetron in SRI-induced sexual dysfunction. J Clin Psychiatry. 2001;62:469-473.
53. Hirose S, Ashby CR Jr. An open pilot study combining risperidone and a selective serotonin reuptake inhibitor as initial antidepressant therapy. J Clin Psychiatry. 2002;63:733-736.
54. Ostroff RB, Nelson JC. Risperidone augmentation of selective
serotonin reuptake inhibitors in major depression. J Clin Psychiatry. 1999;60:256-259.
55. Bobes J, Garca-Portilla MP, Rejas J, et al. Frequency of sexual
dysfunction and other reproductive side-effects in patients with
schizophrenia treated with risperidone, olanzapine, quetiapine,
Sexual functioning and quality of life in patients in remission from depression – Kennedy and Rizvi
TRUE REMISSION
or haloperidol: the results of the EIRE study. J Sex Marital Ther.
2003;29:125-147.
56. Landén M, Eriksson E, Agren H, et al. Effect of buspirone on
sexual dysfunction in depressed patients treated with selective
serotonin reuptake inhibitors. J Clin Psychopharmacol.1999;19:
268-271.
57. Invernizzi RW, Sacchetti G, Parini S, et al. Flibanserin, a potential antidepressant drug, lowers 5-HT and raises dopamine
and noradrenaline in the rat prefrontal cortex dialysate: role of
5-HT(1A) receptors. Br J Pharmacol. 2003;13:1281-1288.
58. Pyke R, Cotton D, Cunningham L, et al. Flibanserin: a novel
centrally acting agent with potential to treat decreased sexual
desire causing personal distress in women. J Clin Psychiatry. In
press.
59. Baldwin D, Hutchinson J, Donaldson K, Shaw B, Smithers A.
Selective serotonin re-uptake inhibitor treatment-emergent sexual dysfunction: randomized double-blind placebo-controlled parallel-group fixed-dose study of a potential adjuvant compound,
VML-670. J Psychopharmacol. 2008;22:55-63.
60. Bernik M, Vieira AH, Nunes PV. Bethanecol chloride for treatment of clomipramine-induced orgasmic dysfunction in males.
Rev Hosp Clin Fac Med Sao Paulo. 2004;59:357-360.
61. Fava M, Nurnberg HG, Seidman SN, et al. Efficacy and safety of sildenafil in men with serotonergic antidepressant-associated erectile dysfunction: results from a randomized, doubleblind, placebo-controlled trial. J Clin Psychiatry. 2006;67:240246.
62. Nurnberg HG, Hensley PL. Sildenafil citrate for the management of antidepressant-associated erectile dysfunction. J Clin
Psychiatry. 2003;64(suppl 10):20-25.
63. Nurnberg HG, Hensley PL, Heiman JR, Croft HA, Debattista
C, Paine S. Sildenafil treatment of women with antidepressantassociated sexual dysfunction: a randomized controlled trial.
JAMA. 2008;300:395-404.
64. Anghelescu I, Urbanek C. Tadalafil in a patient with treatmentresistant depression and antidepressant polypharmacy. J Clin
Psychopharmacol. 2005;25:191-192.
65. Ashton AK, Weinstein W. Tadalafil reversal of sexual dysfunction caused by serotonin enhancing medications in women. J Sex
Marital Ther. 2006;32:1-3.
66. Clayton A, Keller A, McGarvey E. Burden of phase-specific
sexual dysfunction with SSRIs. J Affect Disord. 2006;91:27-32.
67. Clayton AH, Croft HA, Horrigan JP, et al. Bupropion extended release compared with escitalopram: effects on sexual functioning and antidepressant efficacy in 2 randomized, double-blind,
placebo-controlled studies. J Clin Psychiatry. 2006;67:736-746.
68. Clayton A, Kornstein S, Prakash A, et al. Changes in sexual
functioning associated with duloxetine, escitalopram, and placebo in the treatment of patients with major depressive disorder.
J Sex Med. 2007;4:917-929.
69. Dunn JA, Arakawa R, Greist JH, et al. Assessing the onset
of antidepressant-induced sexual dysfunction using interactive
voice response technology. J Clin Psychiatry. 2007;68:525-532.
70. Kennedy S, Fulton K, Bagby R, et al. Sexual function during
bupropion or paroxetine treatment. Can J Psychiatry. 2006;51:
45-52.
71. Papakostas GI. Tolerability of modern antidepressants. J Clin
Psychiatry. 2008;69:8-13.
72. Cyranowski JM, Frank E, Cherry C, et al. Prospective assessment of sexual function in women treated for recurrent major depression. J Psychiatr Res. 2004;38:267-273.
73. Montejo-González AL, Llorca G, Izquierdo JA, et al. SSRI-induced sexual dysfunction: fluoxetine, paroxetine, sertraline, and
fluvoxamine in a prospective, multicenter, and descriptive clinical study of 344 patients. J Sex Marital Ther. 1997;23:176-194.
74. Thase ME, Clayton AH, Haight BR, et al. A double-blind comparison between bupropion XL and venlafaxine XR: sexual functioning, antidepressant efficacy, and tolerability. J Clin Psychopharmacol. 2006;26:482-488.
75. Baldwin D, Moreno RA, Briley M. Resolution of sexual dysfunction during acute treatment of major depression with milnacipran. Hum Psychopharmacol. In press.
76. Delgado P, Brannan S, Mallinckrodt C, et al. Sexual functioning assessed in 4 double-blind placebo- and paroxetine-controlled
IN
DEPRESSION: T
HE
ULTIMATE GOAL
trials of duloxetine for major depressive disorder. J Clin Psychiatry. 2005;66:686-692.
77. Liebowitz MR, Manley AL, Padmanabhan SK, et al. Efficacy,
safety, and tolerability of desvenlafaxine 50 mg/day and 100 mg/
day in outpatients with major depressive disorder. Curr Med Res
Opin. 2008;24:1877-1890.
78. Clayton AH, Zajecka J, Ferguson JM, et al. Lack of sexual dysfunction with the selective noradrenaline reuptake inhibitor reboxetine during treatment for major depressive disorder. Int Clin
Psychopharmacol. 2003;18:151-156.
79. Ozmenler NK, Karlidere T, Bozkurt A, et al. Mirtazapine augmentation in depressed patients with sexual dysfunction due to
selective serotonin reuptake inhibitors. Hum Psychopharmacol.
2008;23:321-326.
80. Feiger A, Kiev A, Shrivastava RK, Wisselink PG, Wilcox CS.
Nefazodone versus sertraline in outpatients with major depression: focus on efficacy, tolerability, and effects on sexual function
and satisfaction. J Clin Psychiatry. 1996;57(suppl 2):53-62.
81. Carvajal García-Pando A, García del Pozo J, Sánchez AS, Velasco MA, Rueda de Castro AM, Lucena MI. Hepatotoxicity associated with the new antidepressants. J Clin Psychiatry. 2002;63:
135-137.
82. Thase ME, Haight BR, Richard N, et al. Remission rates following antidepressant therapy with bupropion or selective serotonin reuptake inhibitors: a meta-analysis of original data from 7
randomized controlled trials. J Clin Psychiatry. 2005;66:974-981.
83. Kennedy SH, Eisfeld BS, Dickens SE, et al. Antidepressantinduced sexual dysfunction during treatment with moclobemide,
paroxetine, sertraline, and venlafaxine. J Clin Psychiatry. 2000;61:
276-281.
84. Chen KC, Yang YK, Lee IH, et al. Sexual dysfunction and
physicians’ perception in medicated patients with major depression in Taiwan. Depression and Anxiety. 2007;0:1-7.
85. San L and Arranz B. Agomelatine: a novel mechanism of antidepressant action involving the melatonergic and the serotonergic system. Eur Psychiatry. 2008;23:396-402.
86. Eison AS, Freeman RP, Guss VB, et al. Melatonin agonists
modulate 5-HT2A receptor-mediated neurotransmission: behavioral and biochemical studies in the rat. J Pharmacol Exp Ther.
1995;273:304-308.
87. Kennedy S, Emsley R. Placebo-controlled trial of agomelatine
in the treatment of major depressive disorder. Eur Neuropsychopharmacol. 2006;16:93-100.
88. Loo H, Hale A, D’haenen H. Determination of the dose of
agomelatine, melatonergic agonist and selective 5-HT2C antagonist, in the treatment of major depressive disorder: a placebocontrolled dose range study. Int Clin Psychopharmacol. 2002;17:
239-247.
89. Olie JP, Kasper S. Efficacy of agomelatine, a MT1/MT2 receptor agonist with 5-HT2C antagonistic properties, in major depressive disorder. Int J Neuropsychopharmacol. 2007;10:661-673.
90. Montejo AL, Pieto N, Matias J, et al. Better sexual acceptability of agomelatine compared to paroxetine in healthy male volunteers using the PRSexDQ Scale. J Psychopharmacol. In press.
FONCTION
SEXUELLE ET QUALITÉ DE VIE DES PATIENTS
APRÈS RÉMISSION D’UNE DÉPRESSION
L’
adoption de la rémission comme critère clinique dans le traitement des
troubles dépressifs majeurs (TDM) a permis de souligner l’importance
des conséquences négatives des symptômes résiduels après un traitement antidépresseur, comme le montrent les différences de perception de la
rémission entre les médecins et les patients. Il est donc nécessaire d’élargir les
critères d’évaluation afin de mieux mesurer la qualité de vie des patients qui ont
été traités pour des TDM. Les troubles sexuels sont souvent rapportés comme
symptômes du TDM et sont plus souvent encore la conséquence de nombreux
traitements antidépresseurs actuels. Pour beaucoup de patients dépressifs, la
dysfonction sexuelle entraîne une détérioration de la qualité de vie, une non
observance du traitement puis son arrêt. De nombreux antidotes ont été proposés pour soulager les troubles sexuels dus aux antidépresseurs, mais très peu
peuvent être recommandés sur la base d’études contrôlées randomisées fiables.
La meilleure stratégie consiste donc dans la prescription d’une monothérapie
(un seul antidépresseur) qui respecte la fonction sexuelle.
Sexual functioning and quality of life in patients in remission from depression – Kennedy and Rizvi
MEDICOGRAPHIA, VOL 31, No. 2, 2009 151
TRUE REMISSION
IN
DEPRESSION: THE ULTIMATE GOAL
n emerging body of evidence indicates that
antidepressants have adverse effects on emotional experience in some people. It is now
well known among health care professionals that
antidepressants such as selective serotonin reuptake inhibitors (SSRIs) may cause anxiety and agitation, especially in the early days of administration.
However, it is much less well known that antidepressants are associated with a syndrome of emotional numbing or blunting, often emerging as depression remits, which can be troublesome enough
to lead to noncompliance. We call this phenomenon
“emotional side effects of antidepressants” rather
than “emotional blunting,” because of the broad
range of impacts on emotional functioning that are
seen.
A
Jonathan PRICE, MD
Guy M. GOODWIN, MD PhD
Department of Psychiatry
University of Oxford
The Warneford Hospital
Oxford, UNITED KINGDOM
Emotional blunting
or reduced reactivity
following remission of
major depression
by J. Price and G. M. Goodwin,
United Kingdom
ntidepressants such as the selective serotonin reuptake inhibitors are
used widely to treat major depression. While they have reasonable efficacy, they also produce adverse effects. The best known of these relate
to physical effects like nausea and the impact on sexual function. However,
anecdotally there appears to be a broader impact on emotional experience. This
is usually described as a reduction in sensitivity or a sense of numbing or blunting, which may emerge as depression remits. While it may be confounded by
the residual effects of depression, patients often attribute it to their medication.
We propose that this phenomenon of emotional side effects of antidepressants
requires further study. There is laboratory evidence to relate it under experimental conditions to the processing of emotion. Reduced detection of negative
emotion in the faces of others can be demonstrated in healthy volunteers who
have taken citalopram or reboxetine for just 1 week. This effect may predict the
clinical problems that we see. We have now also developed an assessment scale
expressly for use in clinical studies. The future will be to look at antidepressants
whose profile appears to be less emotionally blunting — like agomelatine and
other medications now in development with novel mechanisms of action—in
well-designed clinical studies that can illustrate the absolute and relative frequency of what is an increasingly troublesome side effect of current treatments.
A
Medicographia. 2009;31:152-156.
(see French abstract on page 156)
Keywords: depression; emotional processing; sexual function; clinical trial;
outcome assessment; SSRI; side effect; pharmacotherapy; antidepressant
www.medicographia.com
Address for correspondence: Professor Guy Goodwin, Department of Psychiatry,
University of Oxford, The Warneford Hospital, Oxford OX3 7JX, UK
(e-mail: [email protected])
152 MEDICOGRAPHIA, VOL 31, No. 2, 2009
The phenomenon
of emotional side effects
Some patients taking antidepressants report that
while they feel less emotional pain than before
commencing their antidepressant medication, they
also experience a restricted range of emotions and,
in particular, cannot get a “normal” emotional response to everyday events that would usually be associated with, for example, joy or sadness. This complaint is prominent on Web sites where patients
may post their views about their medicines, such
as Cybercity and Rxlist. Box 1 provides several examples derived from such Web sites.
The reduced level of positive emotion in patients
taking SSRIs may be linked both physiologically,
and in the minds of patients who complain about
the experience, to reduced sexual interest. This has
been widely described. There is a reduction both in
desire and arousal. As always in studies in depressed
subjects, there is the potential for confounding the
effects (and after-effects) of depression itself with
the drugs employed to treat it.1 However, in the case
of sexual side effects, there is clear evidence from
effects in healthy male volunteers that the SSRI
paroxetine has an important and immediate effect
on sexual function.2 Moreover, a comparison between the sexual function of patients recovering
from depression but treated with venlafaxine (serotonin and norepinephrine reuptake inhibitor; SNRI)
in one group and the non-SNRI/non-SSRI, agomelatine, in the other, clearly demonstrates an adverse
effect of venlafaxine.3 The situation for emotional
side effects as we have defined them is much less
clear. A review of the wider medical literature reSELECTED
ABBREVIATIONS AND ACRONYMS
DSM-IV TR Diagnostic and Statistical Manual of
Mental Disorders - Fourth Edition
Text Revision
LEIS
Laukes Emotional Intensity Scale
OQESA
Oxford Questionnaire of Emotional
Side effects of Antidepressants
SNRI
serotonin and norepinephrine reuptake inhibitor
SSRI
selective serotonin reuptake inhibitor
Emotional blunting or reduced reactivity following remission of major depression – Price and Goodwin
TRUE REMISSION
veals that little formal research has been done on
antidepressant-associated emotional blunting. However, there has been a steady trickle of reports since
1990, when Hoehn-Saric and colleagues reported
dose-dependent apathy, indifference, loss of initiative, and disinhibition in patients on fluvoxamine
or fluoxetine.4 There have, for example, been further
reports of SSRI-induced apathy, indifference, and
reduced motivation in children, adolescents, adults,
and older adults5-9; of inability to cry10; of reduction
in irritability, aggression, and negative affect11; and
of reduced emotional lability resulting from cerebrovascular accident12-14 or other brain injury.15,16
Opbroek and colleagues17 moved the field conceptually by linking the rating of a wider impact of SSRIs
on emotion to effects specifically on sexual function. The Laukes Emotional Intensity Scale (LEIS)
was used in 15 participants meeting criteria for
DSM-IV (Diagnostic and Statistical Manual of Mental Disorders–Fourth Edition) major depression who
reported SSRI-induced sexual dysfunction, and in
a control group of 16 hospital employees. Compared
with controls, the SSRI group reported significant
reductions in 12 of the 18 LEIS items, including
ability to cry, irritation, care about others’ feelings,
sadness, erotic dreaming, creativity, surprise, anger,
expression of their feelings, worry, sexual pleasure,
and interest in sex.
However, the way in which emotional side effects
should be understood, measured, and so definitively investigated has remained uncertain. We have
recently investigated the nature of this phenomenon
by conducting interviews with people who attribute
emotional symptoms to their SSRI.18 This study
provided evidence that some patients taking SSRIs
experienced significant emotional symptoms that
they strongly attributed to their antidepressant, and
that had a demonstrable impact on their functioning and played a role in their decision making about
ongoing antidepressant adherence. These emotional symptoms could be described within six key
themes—general effects on all emotions, reduction
in positive emotions, reduction in negative emotions, emotional detachment, just not caring, and
a changed personality. These themes are expanded,
and examples given, in Box 2.
DEPRESSION: T
HE
ULTIMATE GOAL
“When my depression is bad enough that I am non-functioning or even suicidal, I just take the antidepressant that works best for me whether I get a
flat affect or not. I need the med simply to survive. However, as I start healing from the depressive episode, the time comes when I am not willing to
continue with the flat affect. I guess that is a real sign of healing, actually.
At that point, I’ve had my doctor change my medications to an antidepressant that may not work quite so well for my depression (though it works
enough to keep me functioning and alive!), yet it does allow me to experience real emotions.”
“The only saving grace of antidepressant medication is the fact that it
saved my life at the time (I was suicidal). However, I now consider that a
mixed blessing. I do not wish to continue to live in this permanently
deadened, apathetic state.”
“I don’t feel particularly depressed, but have completely withdrawn and
emotionally cut off from everything. Had I known this was going to happen,
i’d never have taken them in the first place.”
Box 1. Patient descriptions of emotional blunting taken from the Internet.
All rights reserved.
General effects
on all
emotions
Reduced intensity, or even absence of emotions, which
are flattened, numbed, dulled or blanked; thoughts
rather than feelings; difficulty understanding emotions;
emotions feel fake or artificial; improved emotional
control.
“I think that’s because my brain isn’t in a place where
it can feel stuff. I’m on this constant emotional plain
of blank-blank—not happiness but OK-ness”
Reduction
in positive
emotions
Reduced intensity and frequency, of eg, excitement,
enjoyment happiness, love, affection, passion,
enthusiasm.
“I’d be… aware that I was in a situation or doing something that… should make me happy but it would
just have no real effect.”
Reduction
in negative
emotions
Reduced intensity and frequency, of eg, sadness, anger,
aggression, anxiety and worry. Reduced ability to cry.
“A feeling of being depressed was like cycling over cobble
stones and you’re feeling things a bit too intensely and
too sharply … and the sort of flattening out effect [of antidepressants] is something which is cushioning that…”
Emotional
detachment
Detachment or disconnection from the environment;
from the self; and from other people, including
children, partner and friends.
“I am able to comfort and cuddle [my children] but I feel
that like there is no emotion behind it.”
Just not caring
Not caring about self, about others, about responsibilities; apathy; reduced interest and motivation;
disinhibition; thoughts of self-harm.
“I… felt slightly removed from everything… I just left
things, you know,… things didn’t really matter somehow. And it was only really when I came off them
I realised I’d sort of skated over so much in life…
important issues weren’t so important.”
Changed
personality
Aspects of personality are altered or removed; behavior
is out of character.
“It does dull the edge of your responsiveness. So you end
up wondering whether, and since your personality is
made up of emotions, you feel that your personality has
been shifted sideways or been unbalanced somehow.”
Epidemiology
There are no good data to indicate the frequency of
emotional side effects, when they are most frequent
and most problematic, and whether they are more
common with some currently available antidepressants than others. However, it is clear from Internet reports, our clinical experience, and our experience of recruiting patients to research studies,
that emotional side effects are not uncommon, and
that there is a spectrum of experience from mild
side effects through to severe side effects that is the
source of bitter complaint. Some patients report that
they suffered emotional side effects only as they experienced remission, while others report that they
experienced them throughout the period of antidepressant administration. While SSRIs appear to be
most closely associated with emotional side effects,
IN
Box 2. Emotional side effects of selective serotonin reuptake inhibitors: key themes.
Based on data from reference 18.
Emotional blunting or reduced reactivity following remission of major depression – Price and Goodwin
MEDICOGRAPHIA, VOL 31, No. 2, 2009 153
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
other classes of antidepressant such as SNRIs and
mood stabilizers such as lithium salts have also
been described as doing the same thing.
‘numbed’ or ‘blunted’ in some way, lacking some
positive emotions or negative emotions, feeling detached from the world around you, or ‘just not caring’ about things that you used to care about.”
Impact on quality of life
Little is known about the impact of emotional side
effects on quality of life in remission. However, our
recent qualitative study investigated this in a sample of patients with chronic antidepressant use
(median duration 23 months) and modest levels of
depression (median Beck Depression Inventory–II
score 12.5).18 The impact of emotional side effects
on participants’ daily lives varied widely, both in extent and in perceived helpfulness. Some participants
described emotional side effects as being helpful.
For example, the reduction of certain emotional responses, such as anger, aggression, or worry might
benefit personal relationships. Furthermore, emotional detachment and reduced anxiety might enable a person to think more clearly and objectively
about their life situation.
However, some participants were concerned that
blunting of their emotions and, thereby, of their day
to day concerns, might mask or hide problems. This
might prevent them resolving their own emotional issues, prevent them engaging with other problems or issues requiring their attention, and “cover up” who they really were. “Just not caring” had
an unhelpful effect on everyday responsibilities, resulting in financial problems and problems at work
or college. Emotional detachment from family and
reduced emotional responsiveness had an unhelpful impact on family life, and on perceived quality
of parenting. Reduced inspiration, reduced imagination, reduced motivation, and reduced passion for
and enjoyment of creative activities, had adversely
affected some participants’ creativity. In some participants, emotional side effects had led to reduced
sociability. Finally, emotional flattening, emotional detachment from other people, and reduced concern for other people’s needs and feelings had unhelpful effects on relationships within families, with
a significant other, and at work.
Assessment
Clinical assessment
It is likely that emotional side effects are under-reported in clinical populations due to the lack of systematic enquiry by health care professionals. We
recommend that clinicians ask routinely about
emotional side effects when they are assessing progress with antidepressants. This might comprise
asking a broad screening question, and then, if necessary, following up with more specific questions
to characterize the nature and extent of the problem, the extent to which the patient attributes the
problems to their antidepressant, and its contribution to their decision-making regarding ongoing adherence. An effective screening question
might be: “Have you experienced any emotional
side effects from your antidepressant? (prompt if
required…) Emotional side effects are varied, but
might include, for example, feeling emotionally
154 MEDICOGRAPHIA, VOL 31, No. 2, 2009
Scientific assessment: in the laboratory
Changes in emotional processing can also be shown
to accompany the administration of SSRI and SNRI
drugs to healthy volunteers.19 By emotional processing, one means the capacity of subjects to identify either the emotional expression in the faces of
others (a perceptual expression of emotional sensibility) or to remember emotionally charged self-referent words. The SSRI citalopram given to healthy
volunteers for 7 days and the selective noradrenergic drug reboxetine given for the same period of
time both reduced the capacity of volunteers to accurately detect negative emotion in faces presented
to them in an experimental paradigm. There was
even a reduced signal in the amygdala in response
to the same faces presented subliminally.20 This
could imply that these individuals might be less
sensitive socially to the expression of negative emotions by others. While this could be a helpful change
in emotional processing with regard to both anxiety and depression, it could also, when patients have
recovered, lead to a changed sense of the immediacy of experience.
Changes could also be demonstrated in the domain of emotional memory following the administration of antidepressants to healthy volunteers.
In this case, subjects were more able to recall positive than negative adjectives that could refer to
them. This effect may be interpreted as reflecting a
potentially therapeutic change in how emotion is
processed. However, in potential contrast with the
perception of emotion in faces, it would not itself
lead to a predictable change in emotional experience that would be likely to be negative. Therefore
it is possible that one way to screen for the presence or absence of emotional blunting from any
medication would be to compare the impact on a
patient’s immediate judgment of emotional valence
with the impact on how emotionally charged adjectives are recalled from memory. It is a strong hypothesis of the Oxford group that emotional blunting may be associated with the former, while
antidepressant efficacy per se will be associated
with the latter. Findings with the new antidepressant agomelatine, whose novel mechanism
of action comprises melatonergic agonist activity
at MT1 and MT2 receptors and antagonist activity
at serotonergic 5-HT2C receptors, when given for
1 week to healthy volunteers, will be an important test: the further question will be whether
these properties of agomelatine translate as we
would predict into antidepressant efficacy without
emotional blunting. However, to know if this occurs we will require a better measure of the clinical as opposed to the laboratory experience.
Scientific assessment: in the clinic
A sensitive and valid questionnaire measure of this
phenomenon is needed. This would enable research
into the prevalence of the emotional side effects of
Emotional blunting or reduced reactivity following remission of major depression – Price and Goodwin
TRUE REMISSION
antidepressants, and comparison of the extent to
which individual antidepressants are associated
with them. It could be an important outcome measure in clinical trials designed to compare different
products. Such a measure needs to be carefully designed and validated. Unfortunately, the two existing measures that attempt to address this issue appear to lack either careful design or validation.
The first is LEIS,17 a self-report instrument comprising 18 questions asking patients to rate an aspect of their emotional life compared with their
“usual” state according to a 5-point scale (a lot less/
somewhat less/same as usual/somewhat more/a lot
more). This scale is unvalidated, and the single published report of its use in an observational study
gives few details about its development, stating only
that “we systematically questioned patients treated
with SSRIs about their subjective emotional experience before and after treatment,” and that “these
unstructured interviews led to the development of
a rating scale for SSRI-induced emotional blunting.”
The second is the Bell-Shipman Apathy/Emotional Blunting Questionnaire,21 which is described
in the study report as “under development.” This
self-completion questionnaire comprises five questions, each rated on a six point scale from “strongly
disagree” to “strongly agree,” which relate to aspects
of four of the themes we identified — reduction in
positive emotions, reduction in negative emotions,
emotional detachment, and not caring. However,
there are no published details of the development
and selection of these questions, and no validation
work appears to have been carried out.
We have used the results of the careful observational work in our recent study to develop the Oxford Questionnaire of Emotional Side effects of Antidepressants (OQESA), which we have now piloted,
refined, and validated in a sample of over 200 people taking antidepressants. The OQESA comprises
26 questions/statements in three sections. The respondent rates the extent of their agreement with
each statement along a 5-point scale ranging from
disagree to agree, according to their experience
during the past week. Section 1 includes 12 statements relating to the respondent’s experience of
emotional side effects during the past week (eg, “All
my emotions, both ‘pleasant’ and ‘unpleasant,’ are
‘toned down’”). Section 2 includes 8 statements relating to experience of emotional side effects during the past week compared with the respondent’s
experiences before they developed their illness/problem (eg, “Day to day life just doesn’t have the same
emotional impact on me that it did before my illness/problem”). Within the 20 statements in Sections 1 and 2, the 4 dimensions, general reduction
in emotions (GR), reduction in positive emotions
REFERENCES
1. Montejo AL, Llorca G, Izquierdo JA, Rico-Villademoros F. Incidence of sexual dysfunction associated with antidepressant agents:
a prospective multicenter study of 1022 outpatients. J Clin Psychiatry. 2001;62:10-21.
2. Montejo AL, Prieto N, Terleira A, et al. Better sexual acceptability of agomelatine compared to paroxetine in healthy male
volunteers using the PRSexDQ Scale. Eur Neuropsychopharmacol. 2007;17:S349.
3. Kennedy SH, Rizvi S, Fulton K, Rasmussen J. A double-blind
IN
DEPRESSION: T
HE
ULTIMATE GOAL
(RP), emotional detachment from others (ED), and
not caring (NC) are each represented by 5 statements. Finally, Section 3 comprises 5 statements
regarding the extent to which respondents believe
their antidepressant is responsible for their emotional symptoms (eg, “The antidepressant is preventing me from feeling my emotions in some
way”), and a final statement regarding actual or contemplated non-adherence due to emotional side
effects. A total score can be calculated, being the
sum of the scores of the four dimensions, and represents the extent to which the respondent is affected by emotional side effects. If required, the further
attributional dimension can be scored.
An important and, as yet, unanswered question is
the extent to which emotional symptoms attributed to antidepressants are actually manifestations of
residual depression, rather than being emotional
side effects. Our validation data suggest that two of
the four dimensions of the OQESA (RP and NC) may
be closely related to depression as well as antidepressant-associated emotional blunting, whereas
the other two dimensions (GR and ED) are less
closely related to depression. We therefore recommend that, in high quality experimental work investigating emotional side effects of antidepressants,
two subtotals are calculated—RP plus NC, and GR
plus ED.
Conclusion
In summary, there is emerging evidence that one
important adverse effect of SSRIs and related medicines is a negative impact on the processing of emotional experience. This was first described by patients
themselves, and investigators are now catching up
with the complaint by devising improved ways of
measuring it, both experimentally and, perhaps
most critically, clinically, by the use of an innovative scale. There is an appropriate and increasing
emphasis upon the patient experience in judging
the outcome of treatment with any intervention for
mood disorder. In the case of antidepressants, the
potentially negative effect on emotional experience
is easily confounded with the symptoms of depression itself, but is a cause for concern and further research. It remains to be seen as to whether it proves
to be an easy task to first devise the means to better measure the experience and second to develop
treatments that will be effective in treating depression without producing the problem. The test
of this will come from antidepressants with novel
mechanisms of action, one of the first examples of
which will be agomelatine. Acknowledgements. Some of the work described here was
supported by a grant from Servier to G. M. Goodwin.
comparison of sexual functioning, antidepressant efficacy, and
tolerability between agomelatine and venlafaxine XR. J Clin Psychopharmacol. 2008;28:329-333.
4. Hoehn-Saric R, Lipsey J, McLeod D. Apathy and indifference in
patients on fluvoxamine and fluoxetine. J Clin Psychopharmacol.
1990;10:343-345.
5. Barnhart WJ, Makela EH, Lotocha MJ. SSRI-induced apathy
syndrome: a clinical review. J Psychiatr Pract. 2004;10:196-199.
6. Garland EJ, Baerg EA. Amotivational syndrome associated with
Emotional blunting or reduced reactivity following remission of major depression – Price and Goodwin
MEDICOGRAPHIA, VOL 31, No. 2, 2009 155
TRUE REMISSION
IN
DEPRESSION: T
selective serotonin reuptake inhibitors in children and adolescents. J Child Adolesc Psychopharmacol. 2001;11:181-186.
7. Hoehn-Saric R, Harris GJ, Pearlson GD, Cox CS, Machlin SR,
Camargo EE. A fluoxetine-induced frontal lobe syndrome in an
obsessive compulsive patient. J Clin Psychiatry.1991;52:131-133.
8. Reinblatt SP, Riddle MA. Selective serotonin reuptake inhibitor-induced apathy: a pediatric case series. J Child Adolesc
Psychopharmacol. 2006;16:227-233.
9. Wongpakaran N, van Reekum R, Wongpakaran T, Clarke D.
Selective serotonin reuptake inhibitor use associates with apathy
among depressed elderly: a case-control study. Ann Gen Psychiatry. 2007;6:7.
10. Oleshansky M, Labbate L. Inability to cry during SRI treatment. J Clin Psychiatry. 2006;57:593.
11. Knutson B, Wolkowitz OM, Cole SW, et al. Selective alteration
of personality and social behaviour by serotonergic intervention.
Am J Psychiatry. 1998;155:373-379.
12. Andersen G, Vestergaard K, Riis JO. Citalopram for poststroke pathological crying. Lancet. 1993;342:837-839.
13. Nahas Z, Arlinghaus KA, Kotrla KJ, Clearman RR, George MS.
Rapid response of emotional incontinence to selective serotonin
reuptake inhibitors. J Neuropsychiatry Clin Neurosci.1998;10:
453-455.
14. Panzer MJ, Mellow AM. Antidepressant treatment of patho-
HE
ULTIMATE GOAL
logic laughing or crying in elderly stroke patients. J Geriatr Psychiatry Neurol. 1992;4:195-199.
15. McCullagh S, Feinstein A. Treatment of pathological affect:
response for laughter and crying. J Neuropsychiatry Clin Neurosci. 2000;12:100-102.
16. Seliger GM, Hornstein A, Flax J, Herbert J, Schroeder K. Fluoxetine improves emotional incontinence. Brain Inj. 1992;6:
267-270.
17. Opbroek A, Delgado PL, Laukes C, et al. Emotional blunting
associated with SSRI-induced sexual dysfunction. Do SSRIs inhibit emotional responses? Int J Neuropsychopharmacol. 2002;5:
147-151.
18. Price J, Cole V, Goodwin GM. Emotional side-effects of selective serotonin reuptake inhibitors (SSRIs): a qualitative study of
patients’ experiences. Br J Psychiatry. In press.
19. Harmer CJ, Hill SA, Taylor MJ, Cowen PJ, Goodwin GM.
Toward a neuropsychological theory of antidepressant drug action: increase in positive emotional bias after potentiation of norepinephrine activity. Am J Psychiatry. 2003;160:990-992.
20. Harmer CJ, Mackay CE, Reid CB, Cowen PJ, Goodwin GM. Antidepressant drug treatment modifies the neural processing of
nonconscious threat cues. Biol Psychiatry. 2006;59:816-820.
21. Bell S, Shipman M, Bystritsky A, Haifley T. Fluoxetine treatment and testosterone levels. Ann Clin Psychiatry. 2006;18:19-22.
ÉMOUSSEMENT
ÉMOTIONNEL OU RÉACTIVITÉ RÉDUITE
APRÈS RÉMISSION D’UNE DÉPRESSION SÉVÈRE
L
es inhibiteurs sélectifs de la recapture de la sérotonine sont des antidépresseurs largement utilisés dans le traitement de la dépression sévère.
D’une efficacité satisfaisante, ils ont aussi des effets indésirables dont
les plus connus sont des effets physiques comme la nausée et la dysfonction
sexuelle. De façon anecdotique, ces effets semblent également avoir un impact
touchant plus largement le plan émotionnel. Cet impact est généralement décrit comme une réduction de la sensibilité ou une sensation d’engourdissement
ou d’émoussement qui apparaît lorsque la dépression diminue. Bien que parfois
confondus avec les effets résiduels de la dépression, les patients attribuent souvent ces signes à leur traitement. Nous considérons que ces effets secondaires
des antidépresseurs sur l’émotion nécessitent d’être plus amplement étudiés.
Des données expérimentales permettent sous certaines conditions, de les relier
au processus de traitement de l’émotion. Des volontaires sains qui avaient reçu
pendant juste 1 semaine du citalopram ou de la réboxétine ont montré qu’ils
détectaient moins les émotions négatives sur les visages d’autrui. Un tel effet
pourrait être prédictif des problèmes cliniques que nous observons. Nous avons
développé une échelle d’évaluation expressément pour l’usage clinique. L’avenir est donc aux antidépresseurs entraînant moins d’émoussement émotionnel,
possédant de nouveaux mécanismes d’action, comme l’agomélatine et d’autres
médicaments actuellement en développement. Ces médicaments devront faire
l’objet d’études cliniques bien construites capables de mettre en évidence les
fréquences absolues et relatives d’un effet secondaire de plus en plus gênant liés
aux thérapeutiques actuelles.
156 MEDICOGRAPHIA, VOL 31, No. 2, 2009
Emotional blunting or reduced reactivity following remission of major depression – Price and Goodwin
TRUE REMISSION
IN
DEPRESSION: THE ULTIMATE GOAL
Eugene S. PAYKEL
MD, FRCP, FRCPsych, FMedSci
Department of Psychiatry
University of Cambridge
Cambridge, UNITED KINGDOM
Residual symptoms
and relapse in depression
by E. S. Paykel, United Kingdom
Course of depression
fter the introduction of a range of modern
antidepressants in the early 1960s, it was assumed that the longer-term outcome of depression had become benign. However, in the 1980s
and early 1990s it became clear that in many patients this was not the case. After recognition of
this problem, a paper published in 1991 by Frank et
al1 established and formulated operational definitions for some important concepts and distinctions
among various aspects of outcome, based on severity and time intervals. A more recent US paper 2 has
updated the concepts and definitions.
A
esidual symptoms indicating incomplete remission from depression
present an important clinical problem. They occur in up to a third of depressed patients after acute treatment, and they span the typical symptoms of depression, except those of severe depressive disorder. Their most important consequence is a much increased risk of relapse, particularly in the first
year. Residual symptoms point to the need for further acute treatment to produce greater improvement, if possible. They are a strong indication for longer
than usual continuation treatment at an adequate dose, to prevent relapse.
There is now also good evidence for cognitive therapy as an adjunct. Other persistent abnormalities after depression include social dysfunction, dysfunctional attitudes, hypothalamic-pituitary-adrenal (HPA) axis overactivity, shortened
latency of rapid eye movement (REM) sleep, and mood lowering after tryptophan depletion. It is not clear how much some of these are associated with residual symptoms or occur independently, although continuing HPA axis overactivity and shortened REM latency are also associated with increased risk of
relapse. In addition, there is growing evidence for residual symptoms in bipolar
disorder, particularly after bipolar depression, and for increased risk of relapse
when they are present.
R
Medicographia. 2009;31:157-163.
Partial remission and residual symptoms
Missing from the original schema was consideration of an intermediate state, in which remission
might be partial in degree, with residual symptoms,
rather than complete. This has since emerged as being very important, as it has become clear that it
is a key pointer to relapse and recurrence.
(see French abstract on page 163)
Keywords: partial remission; residual symptom; relapse; depression;
prevention
www.medicographia.com
Address for correspondence: Professor Eugene Paykel, Emeritus Professor of Psychiatry,
University of Cambridge, 18b Trumpington Road, Cambridge CB2 8AH, UK
(e-mail: [email protected])
Residual symptoms and relapse in depression – Paykel
The term remission has usually been applied to
short-term achievement of low or absent symptom
levels, representing an end to the immediate episode.
The term recovery has been used to reflect remission beyond this state, persisting for a longer time
period, and being more complete. A further term,
response, has sometimes been used, implying considerable improvement, variously defined, but not
necessarily to remission.
Even before recovery is fully achieved, relapse may
occur. The term relapse has been used in affective
disorders particularly to describe an early return
of the depressive episode after remission, up to
approximately 9 months to a year following the
acute episode. This has been assumed to be a return
of the original illness. This assumption reflects views
that were common in the early days of antidepressants, ie, that the disorder is merely suppressed and
that the underlying disturbance continues until
spontaneous remission occurs. It is difficult to prove
this theoretical distinction, other than by inferring
it from the length of the symptom-free period. The
term recurrence has been reserved for development
of a subsequent episode, assumed to represent a new
episode.
SELECTED
DSM-III-R
REM
STAR*D
ABBREVIATIONS AND ACRONYMS
Diagnostic and Statistical Manual of
Mental Disorders - Third Edition
Revised
rapid eye movement
Sequenced Treatment Alternatives to
Relieve Depression (study)
MEDICOGRAPHIA, VOL 31, No. 2, 2009 157
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
Residual symptoms received relatively little attention before the mid 1990s, but they are evident
in some studies if the detail is examined, and some
aspects were briefly reviewed.3 Indeed, clinical experience had long suggested that many patients
improved only partially after initial treatment, leaving residual symptoms that persisted and fluctuated in the community, causing much disability. Because most studies grouped these patients either
with those who had not remitted or with those who
had relapsed, their proportion had not been very
well documented.
A few studies described them separately. Among
inpatients treated with amitriptyline, approximately one third were found to be complete responders,
partial responders, and nonresponders, respectively.4 In a follow-up study 5 to 4 years of a sample of
female depressives who had responded to initial
treatment with amitriptyline and had been included in a controlled trial of continuation antidepressant therapy and psychotherapy, many were still
found to show moderate or fluctuating symptoms,
corresponding approximately to residual chronicity, although these included some subjects who had
relapsed and then remitted. Occurrence of residual
symptoms was noted in general practice patients
with depression and anxiety,6 and in 38% of elderly
depressives at 1 year, and 20% at 2-4 years.7
A study of ours first published in 1995 drew specific attention to the problem and gave impetus to
an increased subsequent emphasis. In a prospective
longitudinal follow-up of depressed patients treated in Cambridge in the early 1990s,8,9 a sample of
64 depressed patients meeting the Research Diagnostic Criteria for definite primary unipolar major
depression was studied on presentation, and followed to remission or 15 months. Only 6% of the
sample failed to remit to the criterion of 2 months
below definite major depression by this point. However, on examining the findings in more detail, although the majority of remitters scored in the lower ranges of the 17-item Hamilton Rating Scale for
Depression, an important proportion of 32% scored
8 or more on the Hamilton scale, 8 or less being the
criterion proposed by Frank et al1 as indicating full
remission or recovery. They spanned a range from
8 to 18, although they did not satisfy the criteria for
major depression.
We explored further the nature of these residual
symptoms by examining individual symptom ratings. The residual symptoms were those typical of
depression, with ratings at the level of moderate or
greater on the Hamilton scale items of depressed
mood, impairment of work and activities, psychic
anxiety, and genital symptoms. The remaining depressive symptoms were present to at least a mild
degree in most subjects, the exceptions being a
group of symptoms typical of severe depression: late
insomnia, retardation, agitation, hypochondriasis,
weight loss, and loss of insight. A parallel set of
analyses carried out on the Clinical Interview for
Depression,10 which has a wider range of symptom
items, gave similar findings. Depressed mood, guilt,
hopelessness, impaired work and interests, psychic
anxiety, and anorexia were prominent. The remain158 MEDICOGRAPHIA, VOL 31, No. 2, 2009
ing depressive symptoms were present to at least a
mild degree, except for delayed insomnia, retardation, agitation, panic attacks, increased appetite,
and depressed appearance.
We also looked for initial predictors of later residual symptoms upon remission. Using an extensive
set of ratings made at the initial assessment, we
found very few significant predictors, and those that
were found reflected higher initial severity. Patients
with residual symptoms had higher initial scores on
the Clinical Interview for Depression anxiety total
score and on the Hamilton scale 17-item total score.
Life events, social support, and expressed emotion
did not predict residual symptoms. We also examined diagnoses made at the initial interview, using
the DSM-III-R (Diagnostic and Statistical Manual
of Mental Disorders–Third Edition Revised) criteria
for dysthymia. Those patients with residual symptoms were not predominantly previous dysthymics.
Only 11% of those with residual symptoms satisfied
the DSM-III-R criteria for dysthymia, as opposed to
17% of those without residual symptoms. Residual
major depression did not represent a return to dysthymia, but indicated a different phenomenon: persistence of the episode in spite of treatment.
We also examined treatment data, to explore
whether deficient drug treatment might have been
responsible for residual symptoms. This was not the
case. In fact there was a general trend for patients
with residual symptoms to be receiving higher
rather than lower levels of treatment. This would
be expected in a naturalistic follow-up, with good
treatment assignment in practice based on the presence of more symptoms. It does not mean that higher treatment levels would not be beneficial, but does
indicate that the symptoms were not a consequence
of failure to give standard treatment.
More recent studies of residual symptoms have
been reviewed by Fava et al.11 They have been reported both after drug treatment and psychotherapy. Fava et al,12 in a study of their own, reported a
strong relationship between prodromal and residual symptoms. The most common symptoms were irritability and anxiety. The large Sequenced Treatment Alternatives to Relieve Depression (STAR*D)
study,13 which reported higher non-remission rates
for depression than were hitherto thought to occur,
did not use a criterion for partial remission. Residual symptoms also comprise one aspect of the subthreshold depressive symptoms described in a number of long-term longitudinal studies.
There is also another meaning of the term, to refer to minor symptoms occurring below the level of
partial remission. For example, one or more residual symptoms were found to be present in 82% of
elderly depression remitters who achieved a score of
below 8 on the Hamilton depression scale.14 Among
responders to fluoxetine scoring 7 or less on the
Hamilton scale in another study,15 residual symptoms were frequent, particularly sleep disturbance,
fatigue, loss of interest, and guilt. At these severity
levels, the subjects would be below the usual threshold for partial remission. Possibly the majority of
recovered depressives have these symptoms, but
they overlap, probably to a considerable but so far
Residual symptoms and relapse in depression – Paykel
TRUE REMISSION
Residual symptoms and relapse
Following remission, the patients in our original
study 9 were followed for another 15 months. As in
other follow-up studies, there was a high rate of
subsequent relapse, with 40% of subjects relapsing
over the next 15 months. All the relapses occurred
in the first 10 months, giving some support to the
concept of relapse as an early phenomenon that is
distinguished from recurrence later in time.
An important finding emerged when we separated out the subjects with residual symptoms at remission. Among these, 76% relapsed in the next 10
months compared with 25% of subjects without
residual symptoms (Figure 1).8 Residual symptoms
were a key predictor of subsequent relapse.
Other studies have found high relapse rates in
residual depressives.7,16-19 One study 20 found that patients with residual symptoms of depression obtained greater benefit from maintenance antidepressant therapy than those who had completely
recovered. Prien and Kupfer 21 found that relapse
was less common after full remission of at least 16
weeks, a finding on which they based a recommendation that continuation treatment should comprise
at least 4 months of complete remission. In another study, after 9 months, 49% of a Dutch sample
were found to be in full remission and 45% in partial remission.22 The patients with residual symptoms relapsed early, mainly in the 4 months after
remission, while those without these symptoms had
further episodes that occurred at later than 1 year.
Another study reported that major depressives with
residual symptoms relapsed three times faster than
those without.23 Residual symptoms have been found
to be a strong predictor of relapse in primary care
depressives.24 In Spanish depressive outpatients,25
a relapse rate of 67% was found in the 2 years following partial remission, as opposed to 14% after
full remission. One study 26 looked for the best definition of rating scale scores at 3 or 6 months that
could predict later relapse. No precise cut-off score
with good sensitivity and specificity was found, but
the higher the score, the more likely was subsequent
relapse.
There have been fewer studies of whether residual symptoms at remission predict longer-term recurrence, although some of the above studies fused
earlier relapse and later recurrence in their reports.
We27-29 later extended our original follow-up study to
10 years. The subjects with previous residual symptoms spent more time with depressive symptoms
over the follow-up, but not more time at the full
criteria for major depression, and they showed
greater impairment in social adjustment. No significant differences were found between the two groups
in the percentage having a recurrence in the long
term, the mean number of recurrences, re-admissions, or chronic episodes, or clinical global outcome criteria, although there were small differ-
Residual symptoms and relapse in depression – Paykel
DEPRESSION: T
HE
ULTIMATE GOAL
ences toward a worse outcome on these criteria. The
effects of previous residual symptoms tended to decay over time, and more of the subjects achieved
full remission in due course.
In a trial of maintenance imipramine and interpersonal therapy in patients who had achieved stable remission, the level of residual symptoms did
not predict long-term outcome, but subjects with
greater variability of residual symptoms had a higher risk of recurrence.30 In a similar trial in elderly
patients, residual anxiety and residual sleep disturbance independently predicted early recurrence.31
The treatment of residual symptoms
and prevention of relapse
The most important implications of residual symptoms are the impaired future prognosis and the
need for treatment. There are two aspects to the latter: treatment of the residual symptoms themselves
and prophylactic treatment to prevent relapse.
1
0.9
0.8
Proportion relapsing
ill-explored extent, with what is experienced in normality. These minor symptoms are not the subject
of this paper, which concerns more definite incomplete remission.
IN
0.7
0.6
0.5
0.4
0.3
0.2
0.1
0
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
Months between remission and relapse or end of study
Figure 1. Proportion of patients with ( ) and without ( ) residual symptoms relapsing
after remission from depression.
After reference 8: Paykel ES, Ramana R, Cooper Z, Hayhurst H, Kerr J, Barocka A. Residual symptoms
after partial remission: an important outcome in depression. Psychol Med. 1995;25:1171-1180. Copyright
© 1995, Cambridge University Press.
The association with relapse argues strongly that
residual symptoms should be treated vigorously in
order to abolish them, although there is not conclusive proof from controlled trials that abolishing
or diminishing them in vulnerable subjects will lower the risk of relapse. In any case, these are distressing symptoms, associated with poor role function
and impact on families, and they need treatment in
their own right. However, converting partial remission in these subjects into complete remission may
not be easy. There is often a history of treatment with
a variety of antidepressants in succession in the current and previous episodes, with only partial improvement, and with side effects limiting the doses
that can be achieved.
There is little randomized controlled trial evidence regarding the efficacy of different antidepressants and other treatments for residual symptoms,
and tentative recommendations therefore depend
on general principles. As always, when response is
limited, there should be a process of reassessment.
MEDICOGRAPHIA, VOL 31, No. 2, 2009 159
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
A careful history will often reveal the antidepressant that helped most in this episode or previous
episodes, and that might be used in an increased
dose. Care should be taken before concluding that a
dependent personality is preventing full remission;
chronic illness can create a misleading impression
about a personality, which once full remission is
achieved, is revealed to be robust. Assessment of the
current situation may reveal perpetuating factors,
such as family that has adjusted to the chronic illness by substituting for the patient’s roles, so that
these are not available for the patient to return to
without therapeutic work to change family expectations; or a spouse who needs the patient to be ill;
or elements of secondary gain in the patient, such as
compensation or insurance payments.
1
0.9
Proportion not relapsing
0.8
0.7
0.6
0.5
Control group
CT group
0.4
0.3
0.2
0.1
0
0
4
8
12 16 20 24 28 32 36 40 44 48 52 56 60 64 68
Weeks
Figure 2. Relapse prevention to 18 months, with and without 20 weeks’ cognitive
therapy in patients with residual depression maintained on antidepressants.
CT, cognitive therapy.
After reference 35: Paykel ES, Scott J, Teasdale JD, et al. Prevention of relapse in residual depression
by cognitive therapy. A controlled trial. Arch Gen Psychiatry. 1999;56:829-835. Copyright © 1999,
American Medical Association.
If the patient is currently receiving submaximal
doses of an antidepressant, the dose should be increased as far as can be tolerated, up to maximum
permissible levels. If this fails on its own, augmentation with lithium or other augmentation approaches may be useful. If this too fails, a change of
antidepressant may be considered, to a potent one
shown in controlled trials to produce good remission, without side effects. This should be undertaken with caution, since there is a risk of worsening
symptoms or failing to achieve the previous level of
remission, so addition of a second antidepressant
rather than substitution is often better, if it can be
achieved without any limiting side effects. If there
are clear personal or family issues pointing to psychotherapy, this should be added. For cognitive
therapy, there is plentiful controlled trial evidence
of a prophylactic effect, lessening relapse rates. The
evidence that it lessens residual symptoms themselves is less good, but at the least it provides a useful framework helping the patient to manage the
symptoms, and it will often be indicated in any case,
when residual symptoms do not respond to antidepressant treatment alone, for its prophylactic effect.
160 MEDICOGRAPHIA, VOL 31, No. 2, 2009
The second treatment aspect is the prevention of
relapse and recurrence in these patients, whose risk
is substantial. Here the evidence base is strong. A
key aspect is continuation and maintenance medication, the general value of which is supported by
many controlled trials.32 Earlier, a recommendation
was made that continuation treatment should not
be withdrawn until the patient had experienced 4
months free of all symptoms.21 This recommendation for routine circumstances may in fact be too
short in the light of later evidence that the risk of
relapse extends longer than previously thought.33
Nine to 12 months now seems a better length for
routine continuation. However, the presence of
residual symptoms sufficient to indicate incomplete
remission indicates a need for longer continuation.
Treatment should be continued if possible until they
have become of minor degree or completely subsided, probably for at least 18 months. This may entail quite long-term continuation or maintenance.
When treatment is withdrawn, withdrawal should
be slow. If symptoms return or worsen as doses are
reduced, the full dose should be resumed for a
longer period. If breakthrough symptoms recur as
the dose is reduced, this may argue for indefinite
maintenance.
There is also good evidence for the efficacy of
cognitive therapy in reducing relapse rates,34 particularly in the situation of residual symptoms. Seven
controlled trials have shown reduced relapse rates,
including two studies in residual depression.11,35 Our
own study 35 specifically targeted subjects with residual symptoms in spite of antidepressant treatment
as being a relapse-prone group that needed treatment additional to antidepressants. We found that
adding cognitive therapy to full doses of antidepressant continuation and maintenance lowered relapse
rates (Figure 2), and the effect lasted for 3 and a
half years after the end of the cognitive therapy.36
This appeared to be a specific effect on relapse: residual symptom levels themselves were not lowered.
The evidence is now sufficiently strong that cognitive therapy should routinely be used as adjunctive treatment to medication when partial remission
persists without conversion to full remission.
Other residual phenomena
Israel 37 suggested that recovery from depression
should be determined in three domains: symptoms, psychosocial function, and pathophysiological changes. Social dysfunction and disability are
additional important consequences of a depressive
episode. Social adjustment was evaluated longitudinally in the late 1960s in a sample of depressed
women in New Haven, Connecticut, USA, comparing them with a matched group of normal subjects
in the general population.38,39 Widespread impairment was found in the depressed group compared
with normal subjects, extending across all the domains studied, including work, social and leisure
activities, relationships with extended family, marital relationships, and parental function. These deficits remitted more slowly than did depressive symptoms, and in the 2-month time period that included
Residual symptoms and relapse in depression – Paykel
TRUE REMISSION
response and remission, these deficits were still severe. Improvement in some aspects was incomplete
even at 8 months. Work impairment translates to
decreased productivity and absence from employment, producing some indirect economic costs of
depression. The problems associated with parental
role are particularly important, since problems in
parenting and parent-child relationships impact on
development and later adaptation of the next generation.
Residual social dysfunction has since been reported by many other investigators and has been
found to correlate with symptom outcome. Some
of the many studies40-47 have been reviewed by Fava
et al.11 Residual symptoms are associated with increased social dysfunction. In unpublished data derived from our controlled trial of cognitive therapy
in patients with residual symptoms,35 we examined
mean total scores on the Social Adjustment Scale at
20 weeks. Subjects with residual symptoms at 20
weeks and subjects who had relapsed by 20 weeks
both showed worse social adjustment than those
with neither adverse outcome at this point. Whether
impaired social adjustment makes an independent
contribution to prediction of subsequent relapse has
not so far been reported.
A number of biological and neurocognitive measures, reviewed by Bhagwagar and Cowen,48 have
been found to be abnormal in recovered depressives.
Most prominent have been abnormalities of the hypothalamic-pituitary-adrenal axis, including waking
salivary cortisol49 and dexamethasone non-suppression. The latter has been found to predict relapse.
Several studies that followed up patients treated
with tricyclic antidepressants found that dexamethasone non-suppression at the time of discharge
predicted a greater risk of early relapse.50-57 One
study of outpatients58 and two of patients treated
with electroconvulsive therapy59,60 failed to find
this. The enhanced dexamethasone–corticotropinreleasing-hormone test has also been found to predict relapse.61
A second set of persistent biological abnormalities is related to serotonin. The most prominent of
these is a return of depressive symptoms on depletion of tryptophan by a high amino acid drink low
in tryptophan.62 A third group of abnormalities is
sleep-related, specifically, persistent shortened rapid
eye movement (REM) latency.63 Another group of
abnormalities is neurocognitive. Particularly prominent are the dysfunctional attitudes and attributions that occur in depression and that have also
been found to persist after symptomatic recovery.64,65
The relation of these biological and neurocognitive abnormalities to residual symptoms has not
been well studied, although they do appear to occur
with full remission. Neither is there good evidence
that they predict relapse, other than for dexamethasone suppression and REM sleep latency.
Bipolar disorder
This review primarily concerns unipolar disorder.
However, there is a smaller but growing parallel literature regarding bipolar disorder. Two large pros-
Residual symptoms and relapse in depression – Paykel
IN
DEPRESSION: T
HE
ULTIMATE GOAL
pective follow-up studies have found subthreshold
symptoms present for substantial periods between
episodes,66,67 as have a number of smaller earlier
studies reviewed by Morriss,68 who also noted that
these were most commonly residual after an episode.
Two recent studies69,70 found residual symptoms
early after remission to be common. Keller et al71
had earlier described subsyndromal symptoms in
about half of a sample of bipolar patients in a controlled trial of high- or low-dose maintenance lithium. Both of the large studies found these present
for much longer than the periods of major disorder,
and found that depressive symptoms predominated
over hypomania. There has been less examination
of the prediction of major relapses by these symptoms, but three studies69,70,72 found that, when present, these residual or inter-episode symptoms are
strong predictors of relapse and recurrence.
Conclusions:
the nature and significance
of residual symptoms
What can we say regarding the nature of residual
symptoms in depression and their significance?
There are various possibilities. Residual symptoms
might represent persisting illness, the original illness continuing in milder form. Alternatively they
might represent the phenomena preceding and underlying the depressive episode. Two possible aspects of the latter can substantially be discounted:
subjects with residual symptoms are neither liable
to be diagnosed as dysthymic nor, except to a minor
degree, to show more personality abnormality than
those who remit fully.
A third possible underlying phenomenon is that
the residual symptoms could reflect the cognitive
vulnerability of dysfunctional attitudes. However,
the symptoms shown by residual depressives, although they include negative cognitions, are not
limited to these, but include core mood and functional symptoms of depression. These are too wide
to be related easily to a single abnormality of low
self-esteem.
It thus seems likely, given these findings and the
relative lack of association of residual symptoms
with anything else except subsequent relapse, that
the explanation is the first of those given above, persistence of the original disorder and its underlying
neurobiological substrates. The most likely conclusion is that residual symptoms are a manifestation
of a disorder that, in spite of improvement, is still
present—they are evidence that the disorder continues. This is also supported by the tendency of relapses following residual symptoms to occur early.
It reinforces the important practical treatment message: incomplete remission with residual symptoms
indicates a high risk of relapse and a strong need for
continuing treatment. Acknowledgements: This paper is adapted and updated
from Paykel ES. Partial remission, residual symptoms,
and relapse in depression. Dialogues in Clinical Neuroscience. 2008;10:431-437. With permission from the author and Les Laboratoires Servier.
MEDICOGRAPHIA, VOL 31, No. 2, 2009 161
TRUE REMISSION
IN
DEPRESSION: T
HE
ULTIMATE GOAL
REFERENCES
1. Frank E, Prien RF, Jarrett RB, et al. Conceptualization and rationale for consensus definitions of terms in major depressive
disorder. Remission, recovery, relapse, and recurrence. Arch Gen
Psychiatry. 1991;48:851-855.
2. Rush AJ, Kraemer HC, Sackeim HA, et al; ACNP Task Force.
Report by the ACNP Task Force on Response and Remission in
Major Depressive Disorder. Neuropsychopharmacol. 2006;31:
1841-1853.
3. Fawcett J. Antidepressants: partial response in chronic depression. Br J Psychiatry. 1994;165:37-41.
4. Kupfer DJ, Spiker DG. Refractory depression: Prediction of
non-response by clinical indicators. J Clin Psychiatry.1981;42:
307-312.
5. Weissman MM, Prusoff BA, Klerman GL. Personality in the
prediction of long term outcome of depression. Am J Psychiatry.
1978;135:797-800.
6. Ormel J, Oldenhinkel T, Bri1man E, van den Brink W. Outcome of depression and anxiety in primary care: a three wave 3 1/2
year study of psychopathology and disability. Arch Gen Psychiatry. 1993;50:759-766.
7. Brodaty H, Harris L, Peters K, et al. Prognosis of depression in
the elderly: a comparison with younger patients. Br J Psychiatry.
1993;163:589-596.
8. Paykel ES, Ramana R, Cooper Z, Hayhurst H, Kerr J, Barocka
A. Residual symptoms after partial remission: an important outcome in depression. Psychol Med. 1995;25:1171-1180.
9. Ramana R, Paykel ES, Cooper Z, Hayhurst H, Saxty M, Surtees
PG. Remission and relapse in major depression: a two-year prospective follow-up study. Psychol Med. 1995;25:1161-1170.
10. Paykel ES. The clinical interview for depression development,
reliability and validity. J Affect Disord. 1985;9:85-96.
11. Fava GA, Ruini C, Belaise C. The concept of recovery in major
depression. Psychol Med. 2007;37:307-317.
12. Fava GA, Grandi S, Zielezny M, Canestrari R, Morphy MA.
Cognitive behavioral treatment of residual symptoms in primary
major depressive disorder. Am J Psychiatry.1994;151:1295-1299.
13. Rush AJ, Trivedi MH, Wisniewski SR, et al. Acute and longerterm outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry. 2006;163:
1905-1917.
14. Gasto C, Navarro V, Catalan R, Portella MJ, Marcos T. Residual symptoms in elderly major depression remitters. Acta Psychiatr Scand. 2003;108:15-19.
15. Nierenberg AA, Keefe BR, Leslie BC, et al. Residual symptoms
in depressed patients who respond acutely to fluoxetine. J Clin
Psychiatry. 1999;60:221-225.
16. Faravelli C, Ambonetti A, Pallanti S, Pazzagli A. Depressive relapses and incomplete recovery from index episode. Am J Psychiatry. 1986;143:888-891.
17. Georgotas A, McCue RE, Cooper TB, Nagachandran N, Chang
I. How effective and safe is continuation therapy in elderly depressed patients? Factors affecting relapse rate. Arch Gen Psychiatry. 1988;45:929-932.
18. Simons AD, Murphy GE, Levine JL, Wetzel RD. Cognitive
therapy and pharmacotherapy for depression. Sustained improvement over one year. Arch Gen Psychiatry. 1986;43:43-48.
19. Evans MD, Hollon SD, DeRubeis RJ, et al. Differential relapse
following cognitive therapy and pharmacotherapy for depression.
Arch Gen Psychiatry. 1992;49:802-808.
20. Mindham RH, Howland C, Shepherd M. An evaluation of continuation therapy with tricyclic antidepressants in depressive illness. Psychol Med. 1973;3:5-17.
21. Prien RF, Kupfer DJ. Continuation drug therapy for major depressive episodes: how long should it be maintained? Am J Psychiatry. 1986;143:18-23.
22. Van Londen L, Molenaar RP, Goekoop JG, Zwinderman AH,
Rooijmans HG. Three-to-5-year prospective follow-up of outcome
in major depression. Psychol Med. 1998;28:731-735.
23. Judd LL, Akiskal HS, Maser JD, et al. Major depressive disorder: a prospective study of residual subthreshold depressive symptoms as predictor of rapid relapse. J Affect Disord.1998;50:97-108.
24. Lin EH, Katon WJ, Von Korff M, et al. Relapse of depression in
primary care. Rate and clinical predictors. Arch Fam Med.1998;
7:443-449.
25. Pintor L, Gastó C, Navarro V, Torres X, Fañanas L. Relapse of
major depression after complete and partial remission during a
2-year follow-up. J Affect Disord. 2003;73:237-244.
26. Taylor WD, McQuoid DR, Steffens DC, Krishnan KR. Is there
a definition of remission in late-life depression that predicts later relapse? Neuropsychopharmacology. 2004;29:2272-2277.
27. Kennedy N, Abbott R, Paykel ES. Remission and recurrence
of depression in the maintenance era: long-term outcome in a
Cambridge cohort. Psychol Med. 2003;33:827-838.
162 MEDICOGRAPHIA, VOL 31, No. 2, 2009
28. Kennedy N, Abbott R, Paykel ES. Longitudinal syndromal
and sub-syndromal symptoms after severe depression: 10-year follow-up study. Brit J Psychiatry. 2004;184:330-336.
29. Kennedy N, Paykel ES. Residual symptoms at remission from
depression: impact on long-term outcome. J Affect Disord. 2004;
80:135-144.
30. Karp JF, Buysse DJ, Houck PR, Cherry C, Kupfer DJ, Frank E.
Relationship of variability in residual symptoms with recurrence
of major depressive disorder during maintenance treatment. Am
J Psychiatry. 2004;161:1877-1884.
31. Dombrovski AY, Mulsant BH, Houck PR, et al. Residual
symptoms and recurrence during maintenance treatment of latelife depression. J Affect Disord. 2007;103:77-82.
32. Geddes JR, Carney SM, Davies C, et al. Relapse prevention
with antidepressant drug treatment in depressive disorders: a systematic review. Lancet. 2003;361:653-661.
33. Reimherr FW, Amsterdam JD, Quitkin FM, et al. Optimal
length of continuation therapy in depression: a prospective assessment during long-term fluoxetine treatment. Am J Psychiatry. 1998;155:1247-1253.
34. Paykel ES. Cognitive therapy in relapse prevention in depression. Int J Neuropsychopharmacol. 2007;10:131-136.
35. Paykel ES, Scott J, Teasdale JD, et al. Prevention of relapse in
residual depression by cognitive therapy. A controlled trial. Arch
Gen Psychiatry. 1999;56:829-835.
36. Paykel ES, Scott J, Cornwall PL, et al. Duration of relapse
prevention after cognitive therapy in residual depression: followup of controlled trial. Psychol Med. 2005;35:59-68.
37. Israel JA. Remission in depression: definition and initial treatment approaches. J Psychopharmacol. 2006;20(Suppl 3):5-10.
38. Paykel ES, Weissman MM. Social adjustment and depression:
a longitudinal study. Arch Gen Psychiatry. 1973;28:659-663.
39. Weissman MM, Paykel ES. The Depressed Woman: A study of
Social Relationships.Chicago, Il: University of Chicago Press;1974.
40. Bauwens F, Tray A, Pardoen D, Van der Elst M, Mendlewicz J.
Social adjustment of remitted bipolar and unipolar outpatients.
Brit J Psychiatry. 1991;159:239-244.
41. Goering PN, Lancee WJ, Freeman SJJ. Marital support and
recovery from depression. Br J Psychiatry. 1992;160:76-82.
42. Coryell W, Scheftner W, Keller MB, Endicott J, Maser J, Klerman GL. The enduring psychosocial consequences of mania and
depression. Am J Psychiatry. 1993;150:720-727.
43. Shapira B, Zislin J, Gelfin Y, et al. Social adjustment and selfesteem in remitted patients with unipolar and bipolar affective
disorder. Compr Psychiatry. 1999;40:24-30.
44. Papakostas GI, Petersen T, Denninger JW, et al. Psychosocial
functioning during the treatment of major depressive disorder
with fluoxetine. J Clin Psychopharmacol. 2004;24:507-511.
45. Nasser EH, Overholser JC. Recovery from major depression.
Acta Psychiat Scand. 2005;111:125-132.
46. Agosti V. Predictors of persistent social impairment among
recovered depressed outpatients. J Affect Disord.1999;55:215-219.
47. Furukawa TA, Takeuchi H, Hiroe T, et al. Symptomatic recovery and social functioning in major depression. Acta Psychiat
Scand. 2001;103:257-261.
48. Bhagwagar Z, Cowen PJ. ‘It's not over when it’s over’: persistent neurobiological abnormalities in recovered depressed patients. Psychol Med. 2008;38:307-313.
49. Bhagwagar Z, Hafizi S, Cowen PJ. Increase in concentration
of waking salivary cortisol in recovered patients with depression.
Am J Psychiatry. 2003;160:1890-1891.
50. Goldberg IK. Dexamethasone suppression test as indicator of
safe withdrawal of antidepressant therapy. Lancet. 1980;1:376.
51. Greden IF, Albala AA, Haskett RF, et al. Normalization of dexamethasone suppression test: a laboratory index of recovery from
endogenous depression. Biol Psychiatry. 1980;15:449-458.
52. Holsboer F, Liebl R, Hofschusler E. Repeated dexamethasone
suppression test during depressive illness: normalisation of test
result compared with clinical improvement. J Affect Disord.1982;
4:93-101.
53. Nemeroff CB, Evans DL. Correlation between the dexamethasone suppression test in depressed patients and clinical response. Am J Psychiatry. 1988;141:247-249.
54. Schweitzer I, Maguire KP, Gee AI, Tiller JW, Biddle N, Davies
B. Prediction of outcome in depressed patients by weekly monitoring with the dexamethasone suppression test. Br J Psychiatry.
1987;151:780-784.
55. Charles GA., Schittecatte M, Rush A1, Panzer M, Wilmotte J.
Persistent cortisol non-suppression after clinical recovery predicts symptomatic relapse in unipolar depression. J Affect Disord.
1989;17:271-278.
56. Targum SD. Persistent neuroendocrine dysregulation in
major depressive disorder: a marker for early relapse. Biol Psy-
Residual symptoms and relapse in depression – Paykel
TRUE REMISSION
chiatry. 1984;19:305-317.
57. Yerevanian B, Privitera M, Milanese E, Sagi I, Russotto J. The
dexamethasone test during major depressive episodes. Biol Psychiatry. 1984;19:407-412.
58. Peselow ED, Baxter N, Rieve RR, Barouche F. The dexamethasone suppression test as a monitor of clinical recovery. Am J Psychiatry. 1987;144:30-35.
59. Coryell W. DST abnormality as a predictor of course in major
depression. J Affect Disord. 1990;19:103-109.
60. Katona CLE, Aldridge CR, Roth M, Hyde J. The dexamethasone suppression test and prediction of outcome in patients receiving ECT. Brit J Psychiatry. 1987;150:315-318.
61. Zobel AW, Nickel T, Sonntag A, Uhr M, Holsboer F, Ising M.
Cortisol response in the combined dexamethasone/CRH test as
predictor of relapse in patients with remitted depression. A prospective study. J Psychiatr Res. 2001;35:83-94.
62. Ruhé HG, Mason NS, Schene AH. Mood is indirectly related
to serotonin, norepinephrine and dopamine levels in humans: a
meta-analysis of monoamine depletion studies. Mol Psychiatry.
2007;12:331-359.
63. Giles DE, Jarrett RB, Rush AJ, Biggs MM, Roffwarg HP. Prospective assessment of electroencephalographic sleep in remitted
major depression. Psychiatry Res. 1993;46:269-284.
64. Eaves G, Rush AJ. Cognitive patterns in symptomatic and
remitted unipolar major depression. J Abnorm Psychol.1984;93:
31-40.
SYMPTÔMES
IN
DEPRESSION: T
HE
ULTIMATE GOAL
65. Timbremont B, Braet C. Cognitive vulnerability in remitted
depressed children and adolescents. Behav Res Ther. 2004;42:
423-437.
66. Judd LL, Akiskal HS, Schettler PJ, et al. The long-term natural history of the weekly symptomatic status of bipolar I disorder.
Arch Gen Psychiatry. 2002;59:530-537.
67. Paykel ES, Abbott R, Morriss R, Hayhurst H, Scott J. Sub-syndromal and syndromal symptoms in the longitudinal course of
bipolar disorder. Br J Psychiatry. 2006;189:118-123.
68. Morriss R. Clinical importance of inter-episode symptoms in
patients with bipolar affective disorder. J Affect Disord. 2002;72:
3-13.
69. Tohen M, Bowden CL, Calabrese JR, et al. Influence of sub-syndromal symptoms after remission from manic or mixed episodes.
Br J Psychiatry. 2006;189:515-519.
70. Perlis RH, Ostacher MJ, Patel JK, et al. Predictors of recurrence in bipolar disorder: primary outcomes from the Systematic
Treatment Enhancement Program for Bipolar Disorder (STEPBD). Am J Psychiatry. 2006;163:217-224.
71. Keller MB, Lavori PW, Kane JM, et al. Subsyndromal symptoms in bipolar disorder. A comparison of standard and low serum
levels of lithium. Arch Gen Psychiatry. 1992;49:371-376.
72. Judd LL, Schettler PJ, Akiskal HS, et al. Residual symptom
recovery from major affective episodes in bipolar disorders and
rapid episode relapse/recurrence. Arch Gen Psychiatry. 2008;65:
386-394.
RÉSIDUELS ET RECHUTE DANS LA DÉPRESSION
es symptômes résiduels, signe d’une rémission incomplète de la dépression, sont un important problème clinique. Ils surviennent chez environ
un tiers des patients déprimés après un traitement aigu et ils englobent
les symptômes typiques de la dépression à l’exception de ceux des troubles dépressifs majeurs. Leur conséquence la plus importante est une augmentation
du risque de récidive, surtout la première année. Les symptômes résiduels sont
révélateurs de la nécessité de poursuivre le traitement aigu pour davantage
d’amélioration, si possible, de façon plus prolongée et à une posologie appropriée afin de prévenir une rechute. La thérapie cognitive est maintenant reconnue comme un traitement adjuvant efficace. D’autres anomalies persistantes
sont retrouvées après une dépression comme l’inadaptation sociale, les comportements dysfonctionnels, l’hyperactivité de l’axe hypothalamo-hypophyso-surrénalien (HHS), le raccourcissement du temps de latence du sommeil paradoxal et la baisse de l’humeur après déplétion en tryptophane. On ne sait pas
encore exactement jusqu’à quel point certaines de ces anomalies sont le reflet
de symptômes résiduels ou des manifestations indépendantes, mais l’ hyperactivité de l’axe HHS et le raccourcissement du temps de latence du sommeil paradoxal semblent bien être associés à l’augmentation du risque de récidive. Par
ailleurs, on met de plus en plus en évidence l’existence de symptômes résiduels
dans les troubles bipolaires, en particulier la dépression bipolaire, et leur présence témoigne également de l’augmentation du risque de récidive.
L
Residual symptoms and relapse in depression – Paykel
MEDICOGRAPHIA, VOL 31, No. 2, 2009 163
C
O N T R O V E R S I A L
Q
U E S T I O N
Is the patient really the same
after a major depressive episode?
1
M. Jarema, Poland
epression is a devastating disease for both
the individual and his/her environment.
The simple fact that someone suffers from
depression constitutes a certain danger to the
person in itself. The individual will experience
bothersome symptoms such as lowered mood, loss
of drive, anhedonia, pessimistic thoughts, overwhelming sadness, lack of perspective, low selfesteem, and many other symptoms, causing the
patient’s life to become unbearable and useless.
Clearly, there is an impact on the patient’s life
and his/her family life. Therefore, depression is
often considered as a disorder that affects the
whole family. The most striking example may be
the impact of a suicide attempt on family life.
Thus, after a depressive episode, the patient themself, as well as the family, have experienced a very
unpleasant mental condition that causes them
to fear the next depressive episode. It is clear that
everybody struggles to avoid the next episode or
even the next hospitalization.1 The experience of
a depressive episode seems also not to be neutral
with regard to the patient’s environment. Those
who have survived depression may recognize a
change in the attitudes of other people toward
them; depressive patients may be considered by
others as strange, different, unpredictable, or individuals of a lower vital potential. Studies have
revealed that patients with depression experience
stigmatization like that of patients with schizophrenia.2 Another problematic consequence of a
depressive episode is of a purely biological nature.
Several neurobiological mechanisms underlay
the pathology of depressive symptoms (eg, neuroanatomical and neuroendocrine alterations,
changes in neurotransmitter systems, etc).3 One
can presume that sequential depressive episodes
may cause the hypothetical biological background
D
Marek JAREMA, MD
Professor and Chairman
3rd Department of Psychiatry
Institute of Psychiatry
and Neurology
Ul. Sobieskiego 9, 02-957
Warsaw, POLAND
(e-mail: [email protected])
REFERENCES
1. Harvey PD. Outcomes to monitor when treating bipolar
disorder or schizophrenia. J Clin Psychiatry. 2006;67:e06.
2. Angermeyer MC, Beck M, Dietrich S, Holzinger A. The stigma of mental illness: patients’ anticipations and experiences.
Int J Soc Psychiatry. 2004;50:153-162.
3. Foster JA, MacQueen G. Neurobiological factors linking personality traits and major depression. Can J Psychiatry. 2008;
53:6-11.
164 MEDICOGRAPHIA, VOL 31, No. 2, 2009
of depression to become even more severe, and
each depressive episode may have a certain consequence for the brain as far as biological damage may be concerned. The nature of such consequences may include not only the real “toxic”
influence of the biological factors that presumably constitute the basis of depression, but also
the impact of the biological treatment. Biological
therapy is a first-choice treatment for depression,
but one needs to realize that such a treatment is
under no condition free of any unfavorable impact on the functioning of the brain. The question
indeed may be raised as to why we insist on treating depressive patients with predominantly biological methods, if the expected remission rates
after pharmacological interventions in depression
are only about 50% to 55%? 4 The issue of a negative impact of treatment methods on the patient’s
brain should be given even more attention when
dealing with patients who do not respond adequately to the treatment; the prevalence of “treatment-resistant” depression may constitute 50%
of real-world patients.5 So-called “treatment-resistant patients” are usually subjected to numerous
therapeutic attempts, and many such attempts
are simply ineffective, necessitating introduction
of the next treatment. Clinical experience tells us
that such treatment-resistant patients are difficult to manage when the next depressive episode
appears, because the previous treatment history
causes both the patient and the clinician to develop treatment strategies that include more intensive methods of treatment (ie, higher doses,
drug combinations, augmentation strategies, etc).
Thus, in addition to biological treatment, psychosocial interventions (cognitive behavioral therapy,
emotion-focused therapy, self esteem therapy)6
should be recommended. 4. Huynh NN, McIntyre RS. What are the implications of the
STAR*D trial for primary care? A review and synthesis. Prim
Care Companion J Clin Psychiatry. 2008;10:91-96.
5. Kennedy SH, Giacobbe P. Treatment resistant depression—
advances in somatic therapies. Ann Clin Psychiatry. 2007;19:
279-287.
6. Lau MA. New developments in psychosocial interventions
for adults with unipolar depression. Curr Opin Psychiatry.
2008;21:30-36.
Is the patient really the same after a major depressive episode?
CONTROVERSIAL QUESTION
2
M. Bauer, Germany
ajor depressive disorder (MDD) is a severe mood disorder characterized by
single or recurrent major depressive
episodes. A total of 50% to 85% of patients who
have a depressive episode will eventually go on to
have at least one other episode. Between 9% and
24% of patients with the initial diagnosis of a
major depressive episode will undergo a change
in diagnosis over time, mostly to bipolar disorder.1 Several risk factors have been identified for
depressive relapse. The more episodes that an
individual has, the more likely it is that another
will occur. Other important risk factors include
preexisting dysthymia, a family history of mood
disorders, concurrent anxiety or substance abuse,
and long duration of severe index episode. In recent years it has become apparent that the longterm course of unipolar MDD is not only characterized by high rates of recurrence, but also
dominated by prolonged symptomatic chronicity.2
Many patients with MDD return to their premorbid level of functioning between episodes of major
depression. However, in approximately 30% of
severe or hospitalized depressed patients, residual symptoms and social or occupational impairment persists. Having residual symptoms or subsyndromal depression after acute treatment is
also an important risk factor for relapse and recurrence. About one third of patients suffering
from severe major depression with residual symptoms will have a chronic course marked by at
least 2 years of illness. Epidemiologic and prospective clinical follow-up studies have also documented that the typical course of unipolar MDD involves fluctuating symptoms, whereby depressive
subtypes included in official diagnostic systems
do not represent discrete disorders, but are stages
along a dimensional continuum (spectrum) of
symptomatic severity.3 The group of chronic depressive disorders encompasses four subtypes of
depressive illness: (i) MDD, recurrent, without full
inter-episodic recovery (incomplete remission);
(ii) MDD, currently in a chronic (duration of 2
years) episode (chronic MDD); (iii) dysthymic
disorder; and (iv) “double depression” (concurrent
dysthymic disorder and major depression). The
group of “subthreshold depressions” (depressive
disorders not otherwise specified) includes depressive conditions in which the number, duration, or
quality of symptoms is insufficient to meet the
Diagnostic and Statistical Manual of Mental Disorders criteria for a diagnosis of major depression. Patients with an early onset and older adults
suffering an initial depressive episode after the
age of 60 years appear to be at greater risk for the
development of chronicity. Individuals suffering
from either dysthymia alone or “double depression” have significantly greater impairment in
functioning than those who present with major
depression alone, depressive symptoms, or past
episodes of major depression. Residual (subthreshold) symptoms in the course of MDD are associated with high risk of an early episode relapse
and a significantly more chronic future course of
illness. Recovery from MDD with full resolution
of symptoms is associated with significant delays
in episode relapse and recurrence, and a more
benign course of illness.4 MDD is also associated
with considerable morbidity and mortality, and
for many, an initial episode of depression evolves
into a debilitating chronic illness with significant
and pervasive impairments in psychosocial functioning.5 Chronic depression is characterized by
the increased utilization of health care services,
reduced employment, and greater economic costs
associated with affected individuals. Studies investigating the effects of depression on health-related quality of life demonstrate decrements that
equal or exceed those of patients with chronic
medical illnesses such as ischemic heart disease
or diabetes mellitus.6 Patients who have experienced major depressive episodes are less likely to
sustain a demanding job or career or to achieve
full intellectual potential. If the first depressive
episodes arise during adolescence or early adulthood, diminished performance at school or during
vocational training or university is a frequent
negative outcome with lifelong consequences.
With the information given here, it becomes clear
that the patient is not the same after he or she
has suffered a major depressive episode, because
of the persistent (high) risk of relapse and recurrence and development of a chronic course
of subthreshold depression. Therefore it is important to note that one of the most important
goals in clinical practice is to start with effective
treatment early on, to achieve remission in the
acute phase of treatment, and to attempt as much
as possible to remove all residual depressive
symptoms.7 REFERENCES
1. Angst J, Preisig M. Course of a clinical cohort of unipolar,
bipolar and schizoaffective patients. Results of a prospective
study from 1959 to 1985. Schweiz Arch Neurol Psychiatr. 1995;
146:5-16.
2. Judd LL, Akiskal HS, Maser JD, et al. A prospective 12-year
study of subsyndromal and syndromal depressive symptoms
in unipolar major depressive disorders. Arch Gen Psychiatry.
1998;55:694-700.
3. Angst J, Sellaro R, Merikangas KR. Depressive spectrum
diagnoses. Compr Psychiatry. 2000;41(suppl 1):39-47.
4. Judd LL, Paulus MJ, Schettler PJ, et al. Does incomplete
recovery from first lifetime major depressive episode herald a
chronic course of illness? Am J Psychiatry. 2000;157:1501-1504.
5. Klerman GL, Weissman MM. The course, morbidity, and
costs of depression. Arch Gen Psychiatry. 1992;49:831-834.
6. Wells KB, Stewart A, Hays RD, et al. The functioning and
well-being of depressed patients. Results from the Medical
Outcomes Study. JAMA. 1989;262:914-919.
7. Bauer M, Bschor T, Pfennig A, et al. World Federation of
Societies of Biological Psychiatry (WFSBP) guidelines for biological treatment of unipolar depressive disorders in primary
care. World J Biol Psychiatry. 2007;8:67-104.
M
Michael BAUER, MD, PhD
Professor of Psychiatry
Director, Department
of Psychiatry and
Psychotherapy
University Hospital
Carl Gustav Carus
Technische Universität
Dresden
Fetscherstrasse 74
D-01307 Dresden
GERMANY
(e-mail:
[email protected])
Is the patient really the same after a major depressive episode?
MEDICOGRAPHIA, VOL 31, No. 2, 2009 165
CONTROVERSIAL QUESTION
3
E. Sacchetti, Italy
he wide number of antidepressant drugs
currently available to clinicians constitutes
a valuable opportunity for an improved
management of patients with depression. Nevertheless, the short-term and long-term prognosis
for the disorder remains persistently far from
good in a relevant proportion of patients. Indeed,
among depressed patients, at least one third inadequately responds to the first antidepressant,
and approximately 1 out of 10 remains depressed
even after multiple interventions.1 Furthermore,
residual depressive symptoms are common in
patients who respond to antidepressants.2 Finally,
syndromal and subsyndromal continued depression cause persistent disability and impose a
substantial socioeconomic burden.3,4 Since the
possibility of a true restitutio ad integrum after
depression is nowadays precluded in many patients, the acquisition of new antidepressants with
improved efficacy-safety profiles and mechanisms
of action that are able to work in otherwise refractory patients is certainly welcome. Treatment
response represents indeed the final phenotypic
result of pharmacokinetic and pharmacodynamic
interactions that are both drug- and patient-specific, and are moderated by several other superimposed modifiable and unmodifiable characteristics of the patient, the health care system, wider
society, and/or the environment in general. Two
examples may suffice to support this paradigm:
the first example pertains to poor treatment adherence, a well-known, widespread phenomenon5
that plays a key influential role in the outcome of
depression, because it exposes the individuals to
“false” poor responses, relapses, recurrences, and,
with between-drug differences, withdrawal symptoms. The individual’s propensity to adhere to
therapies for depression is defined by, along with
other variables, cultural and attitudinal factors
in general, social stigma about depression and
its treatment, frequent facilitation by the media
of false conclusions about the utility of conventional antidepressants coupled with overemphasis
on alternative remedies devoid of efficacy, patient
health beliefs, mechanisms of illness denial,
awareness of the need for treatment, decisions
influenced by symptoms, knowledge about antidepressants, previous and current experience
with antidepressants, concerns and aversion for
adverse events, ease and convenience of drug
regimens, cost of medications, and competence
of the treating physician. The second example
refers to the association of residual symptoms
with a longer duration of depressive episode and
increased risk of affective relapse and recurrence.6-10 The persistence of residual symptoms
results not only from the inability of the available therapies to systematically promote a full
recovery, but also on the inappropriate use of
antidepressants and inherent limits in the current
definitions of treatment success. Both these factors indeed promote a partial treatment response,
even when the prescribed treatment has the potential for promoting complete symptom suppression with a return to normal functioning.
Unfortunately, in clinical routine, it is common
to face general practitioners and psychiatrists
who prescribe antidepressants at lower doses and
for shorter periods than are generally recommended by controlled clinical trials, panels of
experts, and international guidelines. In turn, the
conventional guidepost of treatment efficacy
utilized in daily practice is grounded in the reduction of symptom severity at the expense of a
full and sustained remission. Treatment adherence, appropriate prescription of antidepressants,
and use of improved indicators of treatment success may be facilitated by dedicated informative
campaigns, psychoeducational interventions, and
continuous medical education: it seems therefore highly recommendable to spend more time
and resources in these areas of intervention in
order to promote a widespread dissemination
of these proactive strategies that are able to
moderate modifiable modulators of treatment
response. REFERENCES
1. Thase ME, Corya SA, Osuntokun O, et al. A randomized,
double-blind comparison of olanzapine/fluoxetine combination, olanzapine, and fluoxetine in treatment-resistant major
depressive disorder. J Clin Psychiatry. 2007;68:224-236.
2. Cuffel BJ, Azocar F, Tomlin M, Greenfield SF, Busch AB,
Croghan TW. Remission, residual symptoms, and nonresponse
in the usual treatment of major depression in managed clinical practice. J Clin Psychiatry. 2003;64:397-402.
3. Greden JF. The burden of disease for treatment-resistant
depression. J Clin Psychiatry. 2001;62(suppl 16):26-31.
4. Pincus HA, Pettit AR. The societal costs of chronic major
depression. J Clin Psychiatry. 2001;62(suppl 6):5-9.
5. Katon W, Robinson P, Von Korff M, et al. A multifaceted
intervention to improve treatment of depression in primary
care. Arch Gen Psychiatry. 1996,53:924-932.
6. Montgomery SA, Doogan DP, Burnside R. The influence of
different relapse criteria on the assessment of long-term effi-
cacy of sertraline. Int Clin Psychopharmacol. 1991;6(suppl 2):
37-46.
7. Thase ME, Simons AD, McGeary J, et al. Relapse after cognitive behavior therapy of depression: potential implications
for longer courses of treatment. Am J Psychiatry. 1992;149:
1046-1052.
8. Judd LL, Akiskal HS, Maser JD, et al. Major depressive disorder: a prospective study of residual subthreshold depressive
symptoms as predictor of rapid relapse. J Affect Disord. 1998;
50:97-108.
9. Pintor L, Gastó C, Navarro V, Torres X, Fañanas L. Relapse
of major depression after complete and partial remission
during a 2-year follow-up. J Affect Disord. 2003;73:237-244.
10. Kennedy N, Paykel ES. Residual symptoms at remission
from depression: impact on long-term outcome. J Affect Disord. 2004;80:135-144.
T
Emilio SACCHETTI, MD
Professor of Psychiatry
Brescia University
School of Medicine
University Psychiatric Unit
Brescia Spedali Civili,
Piazza Spedali Civili 1
25123 Brescia, ITALY
(e-mail:
[email protected])
166 MEDICOGRAPHIA, VOL 31, No. 2, 2009
Is the patient really the same after a major depressive episode?
CONTROVERSIAL QUESTION
4
P. A. Schmidt do Prado-Lima, Brazil
Pedro Antonio SCHMIDT
DO PRADO-LIMA, MD, PhD
Instituto de Pesquisas
Biomédicas Pontifícia
Universidade Católica do
Rio Grande do Sul (PUCRS)
Rua Alvares Machado, 33-305
Porto Alegre 90 630-010,
BRAZIL
(e-mail: [email protected])
his is a very good question, with a very
complex answer. From any angle that we
examine this matter, it seems that the depressive experience changes the patient. The
amount of information currently available on the
neurobiological toxicity of depression is impressive. A simple search in PubMed using the key
words “depression” and “brain derived neurotrophic factor (BDNF)” produces 517 articles, 89 of them
published in the last 8 months, an example of the
great interest that this topic arouses. The neurobiological approach, which is based mainly on
the observation that patients undergo a decrease
in BDNF and consequently neuronal atrophy and
apoptosis— mostly of CA3 pyramidal hippocampus neurons 1 —establishes the concept of depression-induced brain damage. This brain damage
leads to functional consequences that, for example, predispose the individual to other depressive
reactions when he is facing an adverse environment.1 Depression is also associated with increased
activity of the hippocampus-hypothalamus-pituitary-adrenal (HHPA) axis, due essentially to a
diminished amount and/or sensitivity of glucocorticoid receptors in the hippocampus.2,3 This
adjustment of the axis can persist after a single
depressive episode and can also predispose the
individual to other depressive reactions when he
is confronted by a stressful situation. To illustrate
how a hyperfunctioning HHPA axis can prompt
depressive episodes, we should remember that very
early diminishment of maternal care can lead to
promoter-region methylation of the glucocorticoid
receptor gene in the hippocampus, leading to a
reduction in the expression of this gene and hyperfunctioning of the HHPA axis, which is associated
with post-stress depressive reactions.4 Furthermore, epigenetic modulation through methylation
in the hippocampus is associated with suicide.5 In
an alternative view, using epidemiological data,
we can observe that the occurrence of one depressive episode predisposes the individual to anoth-
er.6 Although this could be due to a genetic predisposition, it is conceivable that the experience
of depression could increase the risk of another
episode.7 I would also like to argue in another
way. Depression is a very intense experience. As
an emotionally charged experience, it deeply influences memory acquisition.8 In other words, the
depressive experience changes the meaning of
many other experiences, since it will be strongly
imprinted. Memory is a form of prolonging the
past; in this way, the consequences of depression
can persist for much longer than the depressive
episode itself. Another memory mechanism can
be influenced by depression. Traditionally, depression has been thought to reduce memory evocation. But is this true? Depressive people remember
negative experiences very well, frequently better
than nondepressive people. This may be explained
by the state-dependent learning concept, proposed
more than 20 years ago.9 To summarize this
concept, the reproduction of the neurochemical
scenario that occurred when the memory was
consolidated facilitates its recall. Of course, during
depression, the negative experiences are what
will be remembered and used in the interpretation
of facts. These negative memories color the daily
events like a negative kaleidoscope. As almost
every new experience is linked to a negative memory, there is a great possibility that they will be
interpreted and stored as being aversive. So, depression leads to the consolidation of strong and
abundant complexes of negative memories about
the self and the environment, which will persist
beyond the depressive episode itself. Interestingly, not every person uses this information in a
negative way. Somehow, some people take advantage of this very aversive experience, using
this negative information in a very positive way.
I think this is the case with some writers and
artists, who transform this overwhelming experience into a productive force.10 But in this case,
depression also changes the person. REFERENCES
1. Duman RS. The neurochemistry of depressive disorders:
preclinical studies. In: Charney DS, Nestler EJ, eds. Neurobiology of Mental Illness. 2nd ed. Oxford, UK: Oxford University
Press; 2004:421-439.
2. Juruena MF, Cleare AJ, Pariante CM. The hypothalamic
pituitary adrenal axis, glucocorticoid receptor function and
relevance to depression. Rev Bras Psiquiatr. 2004:26:189-201.
3. Shelton RC. The molecular neurobiology of depression.
Psychiatr Clin North Am. 2007;30:1-11.
4. Fish EH, Shahrokh D, Bagot R, et al. Epigenetic programming of stress responses through variation in maternal care.
Ann N Y Acad Sci. 2004;1036:167-180.
5. McGowan PO, Sasaki A, Huang TC, et al. Promoter-wide
hypermethylation of the ribosomal RNA gene promoter in
the suicide brain. PLoS ONE. 2008;3:e2085.
6. Mueller TI, Leon AC, Keller MB, et al. Recurrence after
recovery from major depressive disorder during 15 years of
observational follow-up. Am J Psychiatry. 1999;56:1000-1006.
7. Beck AT. The evolution of the cognitive model of depression
and its neurobiological correlates. Am J Psychiatry. 2008;165:
969-977.
8. Izquierdo I, Bevilaqua LRM, Rossato JI, Bonini JS, Medina
JH, Cammarota M. Different molecular cascades in different
sites of the brain control memory consolidation. Trends Neurosci. 2006;29:496-505.
9. Izquierdo I, Netto CA. Role of beta-endorphin in behavioral regulation. Ann N Y Acad Sci. 1985;444:162-177.
10. Schildkraut JJ, Otero A. Depression and The Spiritual in
Modern Art: Homage to Miró. Chichester, UK: John Wiley &
Sons; 1996.
T
Is the patient really the same after a major depressive episode?
MEDICOGRAPHIA, VOL 31, No. 2, 2009 167
CONTROVERSIAL QUESTION
5
H. Ågren, Sweden
ore than a century ago, Emil Kraepelin
divided the major psychoses into dementia praecox (schizophrenia) and manicdepressive illness, using longitudinal progression
and prognosis as major, but not exclusive, dichotomizing concepts. Nevertheless, both Kraepelin
and we know that some schizophrenics get well,
and some manic depressives never will. The concept that depression and mania “always” in time
will heal, has, however, become rooted in clinical
practice. Modern research has nevertheless concluded that if chronicity is defined as never getting out of an episode, then perhaps 1 out of 10
afflicted patients will remain ill for more than
2 years. If chronicity is defined as ever recurring
episodes, then half of patients with an episode
of unipolar depression will experience another
episode, and almost all bipolar patients will continue shifting between mood states if left untreated. So, affective patients get better, but have a
propensity to remain to some extent symptomatic and to suffer recurring episodes. Is major depression, once it has had its debut, a state or a
trait? There is certainly evidence for increasingly
severe episodes: Robert Post developed the kindling or sensitization hypothesis of mood disorders
in the 1980s, stating that episodes tended to accelerate over time and be increasingly independent
of a triggering event,1 and the validity of this concept has been replicated.2 The heavy toll on personal suffering and society has been investigated.3
There is also evidence for neurobiological consequences of mood swings: mood disorders have
been associated with neurodegenerative changes
within the central nervous system—the best researched area being enlarged lateral ventricles and
smaller hippocampi. There is a negative correlation between hippocampal volume and duration
of untreated depression.4 In a recent Anglo-French
report on Diagnostic and Statistical Manual of
Mental Disorders, Fourth Edition (DSM-IV) major
depression, over 8000 outpatients were investigated on two occasions. Cognitive functioning was
analyzed in terms of delayed recall, a memory
function that is particularly related to hippocampal integrity. The study used a structural equation model (path analysis) in showing that the
length of an episode of past depression is correlated with impaired memory performance when the
patient is out of the recent depression, suggesting
a toxic link between the burden of depression and
cognition.5 There is new interest in connecting
inflammation and oxidative stress within the
central nervous system, not only with regard to
dementia and the neurodegenerative disorders,
but also the psychoses and mood disorders.6 Little
is known about the temporal patterns of the central inflammatory process in patients with mood
disorders. With regard to apoptotic cell death within the central nervous system, there is a large
body of evidence for hippocampal neuronal apoptosis in depression7 and the importance of neurogenesis as a mechanism for the therapeutic
effects of antidepressants 8 and electroconvulsive
treatment.9 The presence of neurogenesis in other
areas of the brain, eg, the cortex, remains uncertain. There are steps toward counteracting the
“pessimistic” evidence on depression: (i) there is
general consensus among clinicians about the
value of treating a patient with a mood disorder
to full clinical remission, and the liberal use of
long-term prophylactic treatment, especially (but
not only) in bipolar disorder. Successful relapse
prevention with the use of antidepressant medication is a robust finding—adding another year
of treatment after the initial positive treatment
effect on a depressive episode halves the risk of
relapse.10 The major unresolved issue would rather
be the question of when long-term medication is
unnecessary; (ii) better identification of the origins of environmental risk factors would presumably reduce the risk of relapse or recurrence of
depression.11 An important objective for future
research will be to determine the changes in the
organism that provide the basis for persistence
of the environmental effects on psychopathology.
The best known result of such studies to date has
been the replicated demonstration that White
carriers of two short arms in a polymorphism
within the serotonin transporter gene are more
susceptible to negative life events eliciting depressive episodes in adult life.12 In conclusion, if cerebral alterations in depression are viewed more
as traits than states, then the line between mood
disorders and, for example, schizophrenia would
become blurred. Indeed, recently, genes have been
found that are common to both bipolar disorder
and schizophrenia.13 Few clinical psychiatrists,
however, doubt that mood-stabilizing treatment
helps. Most patients live almost comfortably with
supportive medical and psychological help that
hides their susceptibility to depression, making
them resemble ordinary healthy individuals in
tackling life’s vicissitudes. REFERENCES
1. Post RM, Weiss SRB. Kindling and manic-depressive illness.
In: Bolwig TG, Trimble MR, eds. The Clinical Relevance of Kindling. Chichester, UK: John Wiley & Sons Ltd; 1989:209-230.
2. Ehnvall A, Ågren H. Patterns of sensitisation in the course
of affective illness. A life-charting study of treatment-refractory depressed patients. J Affect Disord. 2002;70:67-75.
3. Kocsis JH, Gelenberg AJ, Rothbaum B, et al. Chronic forms
of major depression are still undertreated in the 21st century:
systematic assessment of 801 patients presenting for treatment.
J Affect Disord. 2008;110:55-61.
4. Sheline YI, Gado MH, Kraemer HC. Untreated depression
and hippocampal volume loss. Am J Psychiatry. 2003;160:
1516-1518.
5. Gorwood P, Corruble E, Falissard B, Goodwin GM. Toxic
effects of depression on brain function: impairment of delayed
recall and the cumulative length of depressive disorder in a
large sample of depressed outpatients. Am J Psychiatry. 2008;
165:731-739.
6. Maes M. The cytokine hypothesis of depression: inflammation, oxidative & nitrosative stress (IO&NS) and leaky gut as
new targets for adjunctive treatments in depression. Neuro
Endocrinol Lett. 2008;29:287-291.
M
Hans ÅGREN, MD, PhD
Professor of Psychiatry
Sahlgrenska Academy,
University of Gothenburg
Institute for Neuroscience and Physiology
Department of Psychiatry
and Neurochemistry
Sahlgrenska University
Hospital/East
SE-416 85 Gothenburg
SWEDEN
(e-mail:
[email protected])
168 MEDICOGRAPHIA, VOL 31, No. 2, 2009
Is the patient really the same after a major depressive episode?
CONTROVERSIAL QUESTION
7. Duman RS, Heninger GR, Nestler EJ. A molecular and
cellular theory of depression. Arch Gen Psychiatry. 1997;54:
597-606.
8. Santarelli L, Saxe M, Gross C, et al. Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science. 2003;301:805-809.
9. Ekstrand J, Hellsten J, Wennström M, Tingström A. Differential inhibition of neurogenesis and angiogenesis by corticosterone in rats stimulated with electroconvulsive seizures. Prog
Neuropsychopharmacol Biol Psychiatry. 2008;32:1466-1472.
10. Geddes JR, Carney SM, Davies C, et al. Relapse prevention
6
with antidepressant drug treatment in depressive disorders:
a systematic review. Lancet. 2003;361:653-661.
11. Rutter M. How the environment affects mental health.
Br J Psychiatry. 2005;186:4-6.
12. Caspi A, Sugden K, Moffitt TE, et al. Influence of life
stress on depression: moderation by a polymorphism in the
5-HTT gene. Science. 2003;301:386-389.
13. Craddock N, O’Donovan MC, Owen MJ. Genome-wide association studies in psychiatry: lessons from early studies of
non-psychiatric and psychiatric phenotypes. Mol Psychiatry.
2008;13:649-653.
J. Pyo Hong, Korea
iverse adverse events such as separation,
physical abuse, and traffic accidents can
occur in everyone’s life and leave sequelae of variable intensity in many areas of human
behavior. Major depressive disorder is a common
but serious condition that causes distress to its
victims, impairs family life, reduces social adjustment, and is a burden on the community.1
Depression is one of the most important public
health problems in Korea: the prevalence of depressive disorders has increased 50% during the
past 5 years according to a recent Korean national epidemiological study,2 and prescriptions of
antidepressants have doubled in the last 5 years.
In addition, the suicide rate in Korea is one of the
highest among the Organization for Economic
Co-operation and Development (OECD) countries.
National depression initiatives from developed
countries emphasize that awareness and recognition of depression is important, because depression is a treatable disease. There are many
antidepressants that have proven efficacy and
safety. However, the past two decades of research
have shown that depression often runs a chronic,
intermittent, lifelong course, although effective
therapeutic approaches have been developed and
provided.3 After acute episodes of illness, many
patients do well for variable periods of time. However, more than half of patients have residual
symptoms and impairment between episodes,
despite ongoing therapy. Anxiety, sleep disturbance, somnolence/fatigue, apathy, and cognitive dysfunction are common residual symptoms
and are associated with an increased risk of relapse and poor psychosocial functioning. Experience of major depressive episodes can lead to a
change in psychological coping styles in patients.
Many patients cope well with depression. They
are able to escape negative thinking, and gain
the capacity to be resilient. However, dynamic
vulnerability theories suggest that consecutive
depressive episodes cause biological and psychological changes that increase the vulnerability
to subsequent depressive episodes. The experience of these episodes reduces the threshold for
the individual’s mind/brain to enter into the depressive state, such that episodes can occur with
little or no environmental precipitant.4 Disabilities related to depression affect work, family, and
social aspects of a patient’s life. Many depressed
patients choose to leave a job because of their
depression and have difficulties in getting a new
job. Due to the negative stigma related to depression, patients often choose not to disclose
their condition at the workplace.5 Research shows
that a significant proportion of men become
depressed when their partners are depressed.
There is an increase in marital conflict and discord within the families of depressed patients.6
Also, many patients with depression are unable
to enjoy social and leisure activities. Alarmingly,
many recovering depressive patients are often
unaware of the long-term consequences of a maladaptive lifestyle, which include dysfunctional
thought, chronic life stress, interpersonal friction, and inadequate rest.7 Major depressive
episodes are believed to leave a substantial impact on many aspects of patients’ lives for a long
time, even after an acute episode has passed.
Development and delivery of innovative psychotherapeutic, psychoeducational, and family therapeutic programs, as well as novel antidepressants, are essential for the recovery of depressed
patients. REFERENCES
1. Klerman GL, Weissman MM. The course, morbidity, and
costs of depression. Arch Gen Psychiatry. 1992;49:831-834.
2. Cho MJ, Hahm BJ, Hong JP, et al. The Epidemiological
Survey of Psychiatric Illness in Korea. Seoul, Republic of Korea: Ministry of Health and Welfare; 2006.
3. Frank E, Kupfer DJ. Progress in the therapy of mood disorders: scientific support. Am J Psychiatry. 2003;160:1207-1208.
4. Kendler KS, Thornton LM, Gardner CO. Genetic risk, number of previous depressive episodes, and stressful life events in
predicting onset of major depression. Am J Psychiatry. 2001;
158:582-586.
5. Depression Alliance. The Inside Story. The Impact of Depression on Daily Life. London, UK: Depression Alliance; 2008.
6. Burke L. The impact of maternal depression on familial
relationships. Int Rev Psychiatry. 2003;15:243-255.
7. Fava GA, Ruini C, Rafanelli C, Finos L, Conti S, Grandi S.
Six-year outcome of cognitive behavior therapy for prevention
of recurrent depression. Am J Psychiatry. 2004;161:1872-1876.
D
Jin PYO HONG, MD
Associate Professor at
Asan Medical Center
University of Ulsan
College of Medicine
388-1 Poonanap-dong
Songpa-gu, Seoul
KOREA
(e-mail:
[email protected])
Is the patient really the same after a major depressive episode?
MEDICOGRAPHIA, VOL 31, No. 2, 2009 169
CONTROVERSIAL QUESTION
7
R. Mahendran, Singapore
he treatment of depression improves a variety of aspects of the illness, such as its symptoms, the patient’s emotional well-being,
and their occupational and social functioning. The
conundrum as to whether the patient is really
the same after a major depressive episode depends
on whether remission or response is achieved.
There are no biological markers for depression at
present. Researchers in clinical trials utilize a
50% improvement on rating scales such as the
Hamilton Rating Scale for Depression (HAM-D)
and the Montgomery-Åsberg Depression Rating
Scale (MADRS) as a guideline for treatment response.1 Although HAM-D is recognized as the gold
standard in clinical trials for depression, it has
been shown that subscales on HAM-D outperform
the total scale in this respect. The available rating
scales do not assess or reflect optimal functioning. In clinical trials, one-third of patients achieve
full remission, one-third achieve a response, and
one-third are nonresponders. Research has shown
that early remission improves long-term wellbeing, and patients treated to remission have fewer
missed workdays. But there are various obstacles
to attaining remission in clinical practice, such
as noncompliance, treatment discontinuation due
to side effects, and failure to recognize residual
symptoms. Many responders may still have a
substantial degree of residual or subthreshold
symptoms, leading to clinically significant negative and unfavorable outcomes.2,3 Paykel et al
found that the rate of relapse in patients with a
partial response was 76%, compared with 25% in
patients achieving full remission.4 Even minimal
levels of residual symptoms contribute to ongoing functional impairment and increase the risk
of further depressive episodes to three to six times
higher than in those with full remission. Additionally, treatment does not automatically improve
social support outcomes. Residual symptoms are
also associated with socioeconomic issues and
help-seeking health care visits, chronicity, and
the risk of suicidal thoughts and attempts.5 Unfortunately, whatever the improvement in mood
symptoms, it is not synchronous with a full return of premorbid capabilities. Zimmerman et al
showed that for patients, psychosocial functioning is of great importance, and positive features
such as “optimism, vigor, and self-confidence”
were crucial in their own assessments of whether
they had returned to their normal selves.6 Patients
who were more likely to view themselves as being in remission reported significantly less psychosocial impairment, and better quality of life.7
Yet findings from studies indicate that treatment
does not ensure immediate and complete recovery of all aspects of the patient’s life. A recent
study found that feelings of hopefulness did not
improve until several weeks or even months after
depressive symptoms were controlled.8 In another study of primary care patients treated with
selective serotonin reuptake inhibitors for major
depression, symptoms continued to improve over
time, while painful physical symptoms persisted.9
In addition, neurobiological investigations of brain
perfusion have shown that there can be a delay
in perfusion normalization for up to 2 years into
remission, with increases in the frontal regions
and decreases in the parietocerebellar regions.10
From the clinician’s perspective, despite the heterogeneity of the etiologic mechanisms underlying depression such as the genetic antecedents,
early trauma, and stress, and the various psychosocial factors such as coping styles, interpersonal
events, and parental bonding experiences, which
can impact recovery, the aim is to have very clear
treatment goals for each patient, with detailed
assessments and monitoring. Attention should be
paid to underlying personality factors, comorbid
psychiatric conditions, and co-existing medical
conditions. The aim is for complete remission, not
only through pharmacotherapy, but also a more
holistic approach with the use of psychotherapeutic treatments and social interventions. REFERENCES
1. Zajecka JM. Treating depression to remission. J Clin Psychiatry. 2003;64(suppl 15):7-12.
2. Casacalenda N, Perry C, Looper K. Remission in major depressive disorder: a comparison of pharmacotherapy, psychotherapy, and control conditions. Am J Psychiatry. 2002;159:
1354-1360.
3. Taylor WD, McQuoid DR, Steffens DC, Ranga Rama Krishnan
K. Is there a definition of remission in late-life depression
that predicts later relapse? Neuropsychopharmacology. 2004;
29:2272-2277.
4. Paykel ES, Ramana R, Cooper Z, Hayhurst H, Kerr J, Barocka A. Residual symptoms after partial remission: an important outcome in depression. Psychol Med. 1995;25:1171-1180.
5. Tranter R, O’Donovan C, Chandarana P, Kennedy S.
Prevalence and outcome of partial remission in depression.
J Psychiatry Neurosci. 2002;27:241-247.
6. Zimmerman M, McGlinchey JB, Posternak MA, Friedman M,
Attiullah N, Boerescu D. How should remission from depression be defined? The depressed patient’s perspective. Am J
Psychiatry. 2006;163:148-150.
7. Zimmerman M, Postemak MA, McGlinchey J, Friedman M,
Attiullah N, Boerescu D. Validity of a self-report depression
symptom scale for identifying remission in depressed outpatients. Compr Psychiatry. 2006;47:185-188.
8. Aikens JE, Kroenke K, Nease DE Jr, Klinkman MS, Sen A.
Psychiatry and primary care. Trajectories of improvement
for six depression-related outcomes. Gen Hosp Psychiatry.
2008;30:26-31.
9. Greco T, Eckert G, Kroenke K. The outcomes of physical
symptoms with treatment of depression. J Gen Inter Med.
2004;19:813-818.
10. Kohn Y, Freedman N, Lester H, et al. Cerebral perfusion
after a 2-year remission in major depression. Int J Neuropsychopharmacol. 2008;11:837-843.
T
Rathi MAHENDRAN, MBBS,
MMed(Psychiatry), FAMS
Chairman, Medical Board
Institute of Mental Health
Buangkok View
Singapore 539747
SINGAPORE
(e-mail: rathi_mahendran@
imh.com.sg)
170 MEDICOGRAPHIA, VOL 31, No. 2, 2009
Is the patient really the same after a major depressive episode?
CONTROVERSIAL QUESTION
8
A. Hatim Sulaiman, Malaysia
oth biological and psychological factors
are believed to play a role in the development of major depressive disorder (MDD).
While the tendency to develop depression may be
inherited or triggered by environmental stressors
such as stressful life events, the traditional view is
that depressive symptoms are a consequence of
an imbalance in the neurotransmitters within the
central nervous system1; namely, norepinephrine,
serotonin, and dopamine, which are involved in
the regulation of mood and emotion. It is well
known that the risk of recurrence increases with
each subsequent episode of depression.2 While the
onset of depressive episodes becomes easily triggered over time, it is progressively less associated
with environmental stressors.3 This phenomenon
supports the “kindling hypothesis,” which proposes that psychosocial stressors play a greater
role in the initial than in subsequent episodes of
depressive disorders.4 Kendler and colleagues, in
their study of white female-female twin pairs, further documented a threshold of approximately
9 episodes of depression at which the brain is no
longer additionally sensitized to the depressive
state.3 Their evidence suggests some neurobiological and neuroanatomical alterations that occur
intensely in the brain during depression, even
during the first few episodes of illness. Chronic
activation of the stress-response system in MDD
can cause a variety of adverse effects in the brain,
and mediates many of the physiological and behavioral responses in depression.5 For example,
an elevated level of corticotropin-releasing factor
in depression is found to mediate some of the
behavioral symptoms of depression involving sleep
and appetite disturbances, reduced libido, and
psychomotor changes. Current evidence has also
shown the involvement of neurogenesis in depression, though its role has not been clearly elucidated yet.1 Neuronal atrophy and cell death occur
as a result of hyperactivity of the stress-response
system, in which adrenal glucocorticoid release
is increased, while brain derived neurotrophic factor (BDNF) levels decline.6 More recent research
suggests an association between depression and
neurogenesis of the hippocampus, a structure
essential to learning and memory, contextual fear
conditioning, and neuroendocrine regulation.7
The damaging effects of prolonged stress could
contribute to the selective loss of hippocampal
volume. A number of neuroimaging studies have
unanimously shown that patients with recurrent
and/or severe depression tend to have smaller
hippocampal volumes.8-11 These morphological
changes were noted to persist long after depressive symptoms had resolved.7 Hippocampal volume reduction is also found to be greater in patients who have had a longer duration of illness
and who went untreated,12-14 indicating that prolonged depression may result in progressive and
cumulative damage to the brain. All these findings underscore the urgency for aggressive treatment early in the course of illness to achieve
remission and to prevent a chronic, recurrent
depressive course. It was demonstrated during a
2-year follow-up study that treatment to the point
of symptom remission in the first 3 months significantly decreased the risk of relapse and recurrence.15 The therapeutic effects of antidepressants are believed to be the result of the drugs’
abilities to restore neurotransmitter imbalance
at the synapses. However, most antidepressants
appear to have a delayed onset of action. An experimental animal model study found that the
onset of therapeutic effects following the initiation
of antidepressant treatment neither coincides
with the achievement of therapeutic serum concentrations nor with the inhibition of neurotransmitters.16 This raises questions about the traditional “chemical imbalance” hypotheses regarding
the underlying pathophysiology of depression.
Additionally, despite the fact that depression is
traditionally viewed as a fully reversible disorder,
accumulating evidence suggests otherwise. More
recent evidence indicates that beyond restoring
the chemical balance at the synapses, antidepressants may have effects associated with re-establishment of neurobiological activity in the brain.
For example, antidepressant pharmacotherapy
affects serotonin and/or norepinephrine activities,
thereby affecting neuronal survival and growth by
decreasing glucocorticoid levels and increasing
BDNF levels.17 In fact, Malberg and colleagues
have shown that chronic antidepressant treatment
was able to increase neurogenesis in the adult rat
hippocampus.18 In human subjects, it has been
reported that patients with mood disorders who
were receiving an antidepressant at their time of
death, had greater hippocampal BDNF expression
compared with untreated subjects.19 In essence,
patients are unlikely to be the same after a major depressive episode, as neurobiological and
neuroanatomical changes have taken place in the
brain. Early and aggressive antidepressant therapy is hence imperative to achieve remission and
prevent recurrence in the long term. REFERENCES
1. Castren E. Is mood chemistry? Nat Rev Neurosci. 2005;6:
241-246.
2. Mueller TI, Leon AC, Keller MB, et al. Recurrence after recovery from major depressive disorder during 15 years of observational follow-up. Am J Psychiatry. 1999;156:1000-1006.
3. Kendler KS, Thornton LM, Gardner CO. Stressful life events
and previous episodes in the etiology of major depression in
women: an evaluation of the “kindling” hypothesis. Am J Psychiatry. 2000;157:1243-1251.
4. Post RM. Transduction of psychosocial stress into the neurobiology of recurrent affective disorder. Am J Psychiatry.
1992;149:999-1010.
5. Arborelius L, Owens MJ, Plotsky PM, Nemeroff CB. The role
of corticotropin-releasing factor in depression and anxiety
disorders. J Endocrinol. 1999;160:1-12.
6. Duman RS, Malberg J, Nakagawa, D’Sa C. Neuronal plasticity and survival in mood disorders. Biol Psychiatry. 2000;48:
732-739.
B
Ahmad HATIM SULAIMAN
MBBS, MPM
Associate Professor
Department of Psychological
Medicine
Faculty of Medicine
University of Malaya
50603 Kuala Lumpur
MALAYSIA
(e-mail: [email protected])
Is the patient really the same after a major depressive episode?
MEDICOGRAPHIA, VOL 31, No. 2, 2009 171
CONTROVERSIAL QUESTION
PROF HATIM SULAIMAN CONTINUED
7. Sapolsky RM. Glucocorticoids and hippocampal atrophy
in neuropsychiatric disorders. Arch Gen Psychiatry. 2000;57:
925-935.
8. Bremner JD, Narayan M, Anderson ER, et al. Hippocampal
volume reduction in major depression. Am J Psychiatry. 2000;
157:115-118.
9. Mervaala E, Fohr J, Kononen M, et al. Quantitative MRI of
the hippocampus and amygdala in severe depression. Psychol
Med. 2000;30:117-125.
10. Steffens DC, Byrum CE, McQuoid DR, et al. Hippocampal
volume in geriatric depression. Biol Psychiatry. 2000;48:301-309.
11. Frodl T, Meisenzahl EM, Zetzsche T, et al. Hippocampal
changes in patients with a first episode of major depression.
Am J Psychiatry. 2002;159:1112-1118.
12. Sheline YI, Gado MH, Kraemer HC. Untreated depression
and hippocampal volume loss. Am J Psychiatry. 2003;160:
1516-1518.
13. Sheline YI, Wang PW, Gado MH, et al. Hippocampal atrophy in recurrent major depression. Proc Natl Acad Sci U. S. A.
9
1996;93:3908-3913.
14. Sheline YI, Sanghavi M, Mintum MA, Gado MH. Depression duration but not age predicts hippocampal volume loss
in medically healthy women with recurrent major depression.
J Neurosci. 1999;19:5034-5043.
15. Simon GE. Long-term prognosis of depression in primary
care. Bull World Health Organ. 2000;78:438-445.
16. Santarelli L, Saxe M, Gross C, et al. Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science. 2003;301:805-809.
17. Gonul AS, Akdeniz F, Taneli F, et al. Effect of treatment on
serum brain-derived neurotrophic factor levels in depressed
patients. Eur Arch Psychiatry Clin Neurosci. 2005;255:381-386.
18. Malberg JE, Eisch AJ, Nestler EJ, Duman RS. Chronic antidepressant treatment increases neurogenesis in adult rat
hippocampus. J Neurosci. 2000;20:9104-9110.
19. Chen B, Dowlatshahi D, MacQueen GM, et al. Increased
hippocampal BDNF immunoreactivity in subjects treated with
antidepressant medication. Biol Psychiatry. 2001;50:260-265.
F. Cañas, Spain
he spontaneous and first answer to this
question should be a clear “no!!,” as any
significant experience in life—and a major
depressive episode undoubtedly is—has the effect
of modifying the subject’s global perspective about
himself and the world. This can be appraised
through the vast literature produced on the matter, one such recent example being W. Styron’s
first person account.1 But this answer probably
fails to respond to the implicit insinuation that
after having recovered from a major depressive
episode, the patient has somehow permanently deteriorated in his functioning (what is known as
the “scarring” effect of depression), and what may
be a consequence of that is a high risk of subsequent episodes. It is well documented that depression frequently has a chronic course with recurrences2 and that the probability of presenting with
a new episode increases with the number of previous ones.3 There are also numerous studies showing that individuals with a history of depressive
episodes have higher scores in some pathological
personality characteristics, such as interpersonal
dependency, lack of social confidence, submissiveness, neuroticism or introversion.4 The role of
these characteristics as being only state markers
or acting as vulnerability factors has been largely
discussed; but a third possibility has emerged
that at least some of them could be a consequence
of the depressive episode, thus producing a kind
of “scar,” a relatively permanent residual deficit
(by analogy with physical scars after an injury).
This is known as the “scar hypothesis” of depression, first introduced as such in the early 1980s.5
Since then, there has been wide controversy
around this possibility, with most published work
on the issue showing results that did not support
the scar hypothesis (it is not possible to quote
them “in extenso,” but a representative sample
may be Rohde et al,6 Shea et al,7 Ormel et al,8 and
Kennedy et al.9 Taken as a whole, they indicate
that postdepressive vulnerabilities correspond to
T
Fernando CAÑAS, MD
Head of Department
of Psychiatry, Hospital
Dr Rodríguez Lafora
Cra de Colmenar
Viejo Km 13.8
28049 Madrid, SPAIN
(e-mail: fcanas.hpma@
salud.madrid.org)
172 MEDICOGRAPHIA, VOL 31, No. 2, 2009
the continuation of premorbid conditions. Nevertheless, some findings are still interpreted as being residual impairments —mostly in the realm
of emotional reactivity—that last longer than the
remission of symptoms and may even become
“latent,”10 so as to mean that they would not appear except in emotionally charged situations.
Almost in parallel to that, increasing attention
has been devoted to the problem of residual symptoms after a depressive episode,11 a phenomenon
whose clinical relevance relates to the dysfunction it can cause and also the risk of relapse that
is significantly increased by its presence. In this
case we will be facing a “badly cured” episode
instead of a permanent scar, with “unresolved
symptoms” that may be under the threshold of
clinical diagnosis, but have a relevance that needs
more detailed study. The prevalence and mechanisms involved in these subthreshold conditions
are not well known,12 but they may force us to
review some of the concepts involved (remission,13
recovery,14 euthymia15), as well as the difficulty
in making prognostic and therapeutic decisions
based upon a diagnostic category so heterogeneous as “major depression,” something that has
already been pointed out.16,17 After more than 50
years of using antidepressant agents, we are still
facing the complexities of the nosology of depression, as it requires an adequate coverage of a
variety of different clinical presentations in terms
of severity and chronicity. Going back to the
initial question, it seems very clear that a major
depressive episode will probably change the patient (although not necessarily for the bad18 if
well treated) but an additional point should also
be that we have to review our standards of treatment, looking for a more ambitious target and
not merely remission of symptoms below the
level of diagnosis.19 If we achieve that, patients
will have a better chance of regaining a level of
functioning that is at least as good as they previously had. Is the patient really the same after a major depressive episode?
CONTROVERSIAL QUESTION
REFERENCES
1. Styron W. Darkness Visible: A Memoir of Madness. New
York, NY: Vintage Books; 1992.
2. Mueller TI, Leon AC, Keller MB, et al. Recurrence after
recovery from major depressive disorder during 15 years of
observational follow-up. Am J Psychiatry. 1999;156:1000-1006.
3. Gonzalez L, Lewinsohn PM, Clarke G. Longitudinal followup of unipolar depressives: an investigation of predictors of
relapse. J Consult Clin Psychol. 1985;53:461-469.
4. Shea MT, Leon AC, Mueller TI, Solomon DA, Warshaw MG,
Séller MB. Does major depression result in lasting personality
change? Am J Psychiatry. 1996;153:1404-1410.
5. Lewinsohn PM, Steinmetz J, Larson D, Franklin J. Depression related cognitions: antecedents or consequences? J Abnorm Psychol. 1981;90:213-219.
6. Rohde P, Lewinsohn PM, Seeley JR. Are people changed by
the experience of having an episode of depression? A further
test of the scar hypothesis. J Abnorm Psychol. 1990;99:264-271.
7. Shea MT, Leon AC, Mueller TI, Solomon DA, Warshaw MG,
Keller MB. Does major depression result in lasting personality change? Am J Psychiatry. 1996;153:1404-1410.
8. Ormel J, Oldehinkel AJ, Vollebergh W. Vulnerability before,
during, and after a major depressive episode. Arch Gen Psychiatry. 2004;61:990-996.
9. Kennedy N, Foy K, Serráis R, McDonough M, McKeon P.
Long-term social functioning after depression treated by psychiatrists: a review. Bipolar Disord. 2007;9:25-37.
10. Merens W, Booij L, Van der Does AJ. Residual cognitive
impairments in remitted depressed patients. Depress Anxiety.
2008;25:E27-E36.
10
11. Paykel ES, Ramana R, Cooper Z, Haydurst H, Kerr J, Barroca A. Residual symptoms after partial remission: an important outcome in depression. Psychol Med. 1995;25:1171-1180.
12. Trivedi MH, Hollander E, Nutt D, Blier P. Clinical evidence
and potential neurobiological underpinnings of unresolved
symptoms of depression. J Clin Psychiatry. 2008;69:246-258.
13. Zimmerman M, Posternak MA, Chelminski I. Heterogeneity among depressed outpatients considered to be in remission.
Compr Psychiatry. 2007;48:113-117.
14. Fava GA, Ruini C, Belaise C. The concept of recovery in
major depression. Psychol Med. 2007;37:307-317.
15. Pizzagalli DA, Goetz E, Ostacher M, Iosifescu DV, Perlis RH.
Euthymic patients with bipolar disorder show decreased reward learning in a probabilistic reward task. Biol Psychiatry.
2008;64:162-168.
16. Parker G. Beyond major depression. Psychol Med. 2005;
35:467-474.
17. Parker GB. Commentary on diagnosing major depressive
disorder. Ask less that we embrace major depression and ask
more what the concept does for us. J Nerv Ment Dis. 2006;194:
155-157.
18. Dahlberg KM, Waern M, Runeson B. Mental health literacy
and attitudes in a Swedish community sample—investigating
the role of personal experience of mental health care. BMC
Public Health. 2008;8:8.
19. Koivumaa-Honkanen H, Tuovinen TK, Honkalampi K, et
al. Mental health and well-being in a 6-year follow-up of patients with depression: assessments of patients and clinicians.
Soc Psychiatry Psychiatr Epidemiol. 2008;43:688-696.
F. T. Antun, Lebanon
ajor depressive illness is a chronic relapsing condition that sometimes warrants lifetime treatment. Studies on the
long-term outcome of major depression have
shown a significant difference between unipolar
and bipolar types. More confusion is added when
unipolar depression switches to bipolarity in later
years, thus affecting the results of long-term outcome studies.1 Furthermore, the classification
and diagnosis of depression is in effect that of a
cluster or set of symptoms, or a syndrome. We
therefore treat these symptoms, and not a diagnosis as such.2 Thus response and quality of life
is measured by the response of these symptoms
to treatment. The diagnostic categories of major
depression and the presence of comorbidities affect the results of studies on the long-term outcome of depressive illness. Antidepressants have
grown in number, as have their different mechanisms of action and molecular types. They all
show varying degrees of ability to generate a
response in comparison with placebo. Furthermore, short-term studies show that all antidepressants have a low advantage over placebo (15%20%) ie, 50% of depressed patients improve on
placebo, although we are unable to know which
patients do well on placebo.3 The duration of
depression treatment in clinical practice varies,
even though treatment algorithms do exist, and
Isomesta et al 4 showed that about 70% of patients
receive subtherapeutic doses of antidepressant.
Randomized controlled trials by Schulberg et al 5
and Katon et al 6 showed that adequate treatment
M
Fuad T. ANTUN, MD, PhD,
FRCP, FRCPsych, DPM
Professor of Psychiatry
Advisor on Mental Health
& WHO Focal Point
Ministry of Health, Lebanon
Chairman, Scientific
Council for the Arab
Board of Psychiatry
PO Box 135098
Beirut, LEBANON
(e-mail:
[email protected])
Is the patient really the same after a major depressive episode?
according to the US Agency for Health Care Policy and Research guidelines gives a clinical outcome that is superior to that of usual treatment.
Gender and the response to antidepressants has
also been studied, and has shown that tricyclic
antidepressants are more effective in males and
that selective serotonin reuptake inhibitors are
more effective in females.7 Compliance is difficult
to study, and is a problem that is not resolved in
assessing outcome.8 Last but not least, cure is a
relative term whose definition varies from symptom relief, to social recovery, adaptive behavior,
and remission. It is infrequently the case that a
patient is cured of depression, in the sense that
no further attacks of depression occur in the lifespan of the patient, and this is even less frequent
with bipolar depression. Against the backdrop of
all the above intricacies, controversies, and clinical variations, there is increasing literature on the
unsatisfactory degree of remission achieved with
current treatments in depression. A Medline review
of relevant articles published from 1967 to 2006
showed that when psychotherapy and pharmacotherapy are used to treat depression, most patients
report residual symptoms despite apparently successful treatment. These residual symptoms have
strong prognostic value, as they have a relationship with prodromal symptoms that can lead to
relapse.9 This subject has received inadequate
attention. Review of the literature using Medline
also showed that some clinical findings point toward: (i) unfavorable long-term outcomes in depression treated by available antidepressants
MEDICOGRAPHIA, VOL 31, No. 2, 2009 173
CONTROVERSIAL QUESTION
(paradoxical depression-inducing effects), with
anxiety and depressive symptoms; (ii) antidepressant-induced switching and cycle acceleration in
bipolar disorders; (iii) occurrence of a tolerance
to antidepressants in long-term treatment; and
(iv) withdrawal effects after stopping treatment,
with possible relapse.10 Residual symptoms are the
rule in unipolar depression. The symptoms are
related to the hypothalamic-pituitary-adrenal
axis and to sleep architecture, and are predictors
of relapse, as they progress to become prodromal
symptoms of relapse upon recovery from the depressive episode. So antidepressants should not
only be assessed on their differential remission
rates, but also on the differential amount of residual symptomatology after response.11 Treatment
of these residual symptoms may improve longterm outcome, so that recovery lies not exclusively in the alleviation of the negative, but also in
engendering the positive. The above discussion
focuses mainly on unipolar depression, in which
there is growing controversy about the concept
of “total cure.” The situation is no better in bipolar depression, where the spectrum of patients
runs the full range from highly functional, welladjusted individuals who are almost completely
symptom-free for decades, to socially dysfunctional, chronically ill patients with repeated relapses,
who never recover.12 In general, in bipolar illness,
depression has a more chronic natural course
than the mania, with patients reporting chronic
subsyndromal depression.13 In conclusion, unipolar depression, a chronic and relapsing illness,
sometimes runs a course that is without full recovery, full recovery being a term that can be confused with remission or social recovery, meaning
the patient is never really the same after a major
depressive episode. There is more controversy
with regard to bipolar depression, where it sometimes takes over 10 years for a diagnosis to be
made, and where the progress toward recovery is
often poor,14,15 with no remitting cyclical course. REFERENCES
1. Akiskal HS, Maser JD, Zeller P, et al. Switching from
unipolar to bipolar II. Arch Gen Psych. 1995;52:114-123.
2. Stefanis CN, Stephanis NC. Diagnosis of depression:
a review. In: Maj M, Sartorius N, eds. Depressive Disorders.
2nd ed. West Sussex, UK: John Wiley & Sons; 2002: Chapter I
(1-151).
3. Bech P. Pharmacological treatment of depressive disorders:
a review. In: Maj M, Sartorius N, eds. Depressive Disorders.
2nd ed. West Sussex, UK: John Wiley & Sons; 2002: Chapter II
(89-117).
4. Isometsa E, Seppala I, Henriksson M, et al. Inadequate
dosaging in general practice of TCA vs other antidepressants
for depression. Acta Psychiatr Scand. 1998:98:451-454.
5. Schulberg HC, Block MR, Madonia MJ, et al. Treating major depression in primary care practice, eight month clinical
outcome. Arch Gen Psych. 1996;53:913-919.
6. Katon W, Robinson P, Vonkorff M, et al. A multifaceted intervention to improve treatment of depression in primary
care. Arch Gen Psych. 1996;53:924-932.
7. Keller MB, Gelenberg AJ, Hirschfeld RMA, et al. The treatment of chronic depression, Part 2. A double-blind randomized
trial of sertraline and imipramine. J Clin Psychiatry. 1998;
59:598-607.
8. Demyttenaere K. Compliance during treatment with antidepressants. J Affect Disord. 1997:43:27-39.
9. Fava GA, Ruini C, Belaise C. The concept of recovery in
major depression. Psychol Med. 2007;37:307-317.
10. Fava GA. Can long-term treatment with antidepressant
drugs worsen the course of depression? J Clin Psychiatry.
2003;64:123-133.
11. Fava GA, Fabbri S, Sonino N. Residual symptoms in depression: an emerging therapeutic target. Prog Neuropsychopharmacol Biol Psychiatry. 2002;26:1019-1027.
12. Lerer B, Yakir A. Heterogeneity of course and outcome in
bipolar disorder: can genetics help? In: Maj M, Akiskal HS,
Lopez-Ibor JJ, Sartorius N, eds. Bipolar Disorder. New York,
NY: John Wiley and Sons. 2002:178-181.
13. Coryell W, Turvey C, Endicott J, et al. Bipolar I affective
disorder: predictors of outcome after 15 years. J Affect Disord.
1998:50:109-116.
14. Golderg JF, Harrow M, eds. Bipolar Disorders: Clinical
Course and Outcome. Washington, DC: American Psychiatric
Press; 1999.
15. Lish JD, Dime-Meenan S, Whybrow PC, et al. The National
Depressive and Manic-Depressive Association (DMDA) survey
of bipolar members. J Affect Disord. 1994;31:281-294.
174 MEDICOGRAPHIA, VOL 31, No. 2, 2009
Is the patient really the same after a major depressive episode?
V
A L D O X A N
BETTER QUALITY REMISSION
IN DEPRESSION: VALDOXAN, THE FIRST
MELATONERGIC ANTIDEPRESSANT
by C. Muñoz, France
espite all the treatments developed in recent
years, depression remains a disabling illness.
Existing antidepressants have mixed clinical
benefits and side effect profiles that often impair
quality of life. It was initially hoped that selective
serotonin reuptake inhibitors (SSRIs) and serotonin norepinephrine reuptake inhibitors (SNRIs)
would circumvent the disadvantages of tricyclic antidepressants (TCAs) and selective or non-selective
monoamine oxidase inhibitors (MAOIs). The reality
is that although SSRIs and/or SNRIs and other dual
action antidepressants are better tolerated, they still
have undesirable side effects. Sexual dysfunction,
gastrointestinal upsets, weight gain, antidepressant
discontinuation syndrome, and sleep disturbance
not only decrease and, in some cases, destroy adherence, they also prevent complete remission. Indeed,
complete and sustained remission has become the
main goal in treating major depressive disorder
(MDD). Incomplete remission is viewed as treatment failure, with residual symptoms being a predictor of relapse and recurrence.1 Hence the need
for novel pharmacologic entities that can offer relief from the illness together with a significant reduction in residual symptoms. Candidates have, for
example, included receptor antagonists to neurokinin 1 (NK1) and corticotropin releasing factor
(CRF).2,3 However, neither class offered consistently positive results in clinical trials and both also act,
D
G
Carmen MUÑOZ, PhD
Division of Medical Information
Servier International
Suresnes, FRANCE
ood quality remission is rarely achieved with conventional antidepressants, suggesting that targets other than the
monoaminergic system hold the key to effective drug therapy. A novel example is circadian rhythm regulation, based on
the finding that circadian rhythms are disturbed in depressed
patients and that such disturbance is an important factor in depressive episode outcome. Valdoxan (agomelatine) is the first
melatonergic antidepressant. As the first antidepressant to be an
agonist at melatonergic MT1 and MT2 receptors and an antagonist at serotonergic 5HT2C receptors, it has a unique receptor
profile for regulating disturbed circadian rhythms. It has demonstrated antidepressant efficacy against placebo in the short and
long term, irrespective of disease severity, and has also outperformed venlafaxine and sertraline on several rating scales, especially those reflecting clinical practice. Efficacy is associated
with unique clinical benefits such as non-sedative regulation of
albeit indirectly, via monoaminergic mechanisms.
A more innovative and promising strategy is to base
antidepressant therapy on the regulation of circadian rhythms. The rationale that resulted in the
synthesis of Valdoxan (agomelatine S 20098) is based
on the observation that circadian rhythms are disturbed in depressed patients4,5 and that abnormal
rhythms play a key role in depressive episode outcome.6 Valdoxan is the first melatonergic antidepressant. Its binding profile differs from that of all
other antidepressants. Valdoxan is a potent melatonergic MT1 and MT2 receptor agonist and a serotonergic 5HT2C receptor antagonist. As such, it
represents a novel pharmacologic approach to the
treatment of MDD.
This review highlights the innovation represented by this mechanism of action, summarizes the
drug’s preclinical and clinical properties, and identifies the features that enable this fresh approach
to the treatment of depression to ensure a better
quality of remission.
Pharmacologic properties of Valdoxan:
a truly innovative antidepressant therapy
Receptor profile
Valdoxan binds strongly to cloned human MT1 and
MT2 receptors with affinity (Ki) values of 0.10 ±0.1
nM and 0.12 ±0.02 nM.7 Its estimated Ki for 5HT2C
the sleep-wake cycle and improved daytime functioning as early
as the first treatment week. Valdoxan does not depress libido or
sexual function, nor induce antidepressant discontinuation syndrome or weight gain, and it is well tolerated. These features make
it a truly novel approach to depression, providing early symptomatic relief within a framework of complete and sustained remission.
Medicographia. 2009;31:175-181.
(see French abstract on page 181)
Keywords: internal clock; circadian rhythm; unique receptor
profile; antidepressant efficacy; remission; discontinuation syndrome; sexual function; body weight; Valdoxan (agomelatine)
www.medicographia.com
Address for correspondence: Carmen Muñoz, Servier International, 35 rue de Verdun,
92284 Suresnes Cedex, France (e-mail: [email protected])
Better quality remission in depression: Valdoxan, the first melatonergic antidepressant – Muñoz
MEDICOGRAPHIA, VOL 31, No. 2, 2009 175
VALDOXAN
Antagonist
5HT2C receptor antagonism. No other increase in
dopamine has been observed in other brain areas,
specifically nucleus accumbens or striatum.
Agonist
5-HT2C receptors
Melatonergic receptors
IC50 1.310–10
MT1
MT2
IC50 2.710–7
IC50 4.710–10
5-HT2C
Negligible affinity for other receptors or transporters
No influence on extracellular serotonin levels
Figure 1. Receptor profile of Valdoxan. Valdoxan is an agonist at melatonergic MT1 /MT2 receptors and an antagonist at 5HT2C receptors.
Its affinity for other receptors or transporter sites is negligible. IC50,
inhibitory concentration–50.
receptors is 710 nM (pKi=6.15 ±0.04).8 Inhibitory
concentration 50 (IC50) values for MT1 , MT2 and
5HT2C receptors are 0.13, 0.47, and 270 nM (Figure 1). Screening of over 80 receptors and enzymes
has revealed no other relevant affinities. Thus unlike the SSRI fluoxetine, Valdoxan (10-50 mg/kg intraperitoneal [IP]) affects neither 5HT1A receptor
density nor basal electrophysiologic characteristics
and responses to 5HT1A receptor stimulation in the
dorsal raphe (presynaptic receptors) or hippocampus (postsynaptic receptors).9 Nor does Valdoxan
(10-40 mg/kg IP) affect serotonin outflow under
acute and chronic conditions.8,10 In freely moving
rats it increases dopamine outflow in frontal cortex
and norepinephrine levels in frontal cortex and the
hippocampus. This effect appears to be mediated by
SELECTED
ABBREVIATIONS AND ACRONYMS
CGI-I
CGI-S
CRF
DESS
HAMA
HAM-D17
5HT
IC 50
IP
LSEQ
MADRS
MAOI
MDD
NK1
PO
PRSEXDQSALSEX
SNRI
SSRI
TCA
TEAE
VAS
Clinical Global Impression–Improvement (scale)
Clinical Global Impression–Severity (scale)
corticotropin releasing factor
Discontinuation Emergent Signs and Symptoms
Hamilton Anxiety Scale
17-item Hamilton Rating Scale for Depression
5-hydroxy tryptamine (serotonin)
inhibitory concentration–50
intraperitoneal
Leeds Sleep Evaluation Questionnaire
Montgomery-Åsberg Depression Rating Scale
monoamine oxidase inhibitor
major depressive disorder
neurokinin-1
per os
Psychotropic-Related Sexual DysfunctionSalamanca Sex Questionnaire
serotonin norepinephrine reuptake inhibitor
selective serotonin reuptake inhibitor
tricyclic antidepressant
treatment-emergent adverse event
Visual Analog Scale
176 MEDICOGRAPHIA, VOL 31, No. 2, 2009
Valdoxan demonstrates efficacy in all animal
models of depression
Valdoxan (10-50 mg/kg per os [PO] or IP) has shown
antidepressant efficacy in a comprehensive and validated battery of animal models11-14: despair test, olfactory bulbectomy, transgenic mice with glucocorticoid receptor deficiency, unavoidable aversive
light stimulus, learned helplessness, and chronic
mild stress. In the latter model, a melatonergic agonist abolishes the antidepressant effect observed in
the evening but not that observed in the morning,
clearly showing that the antidepressant efficacy of
Valdoxan requires both melatonergic and 5HT2C receptors.
Resynchronization of circadian rhythms
Resynchronization in models of circadian
rhythm disturbance
In rats, single or repeated dosing with Valdoxan (510 mg/kg PO) resynchronizes locomotor activity
in models of jet lag, blindness, and delayed-phase
sleep; it also resynchronizes circadian rhythms in
two therapeutic models: aging and trypanosome
infection.15-18
Resynchronization of circadian rhythms in
an animal model of depression
In a naturalistic and well-validated animal model
of depression (the subdominant tree shrew), Valdoxan (40 mg/kg PO) resynchronizes body temperature, while restoring body weight and urinary cortisol.19 Melatonin and 5HT2C antagonists have no
effect in the same model.20
Resynchronization of circadian rhythms in
healthy volunteers
In healthy volunteers, chronic exposure to a therapeutic dose of Valdoxan (50 mg) induced an approximate 2-hour phase advance of body temperature and cortisol rhythm and an increase in plasma
growth hormone. No effect on polysomnography
variables is observed.21 These results are reinforced
by the impact of Valdoxan in depressed patients (see
next section: Clinical studies).
Other activities
Single dosing with Valdoxan (10-40 mg/kg IP) has
proven anxiolytic activity in the elevated plus maze,
social defeat model, Geller-Seifert and Vogel conflict
tests, and social interaction tests. 5HT2C antagonists are effective in these models and melatonin
is effective only in the elevated plus maze test.10,22
Chronic evening dosing with Valdoxan (40 mg/kg IP)
increases cell proliferation, neurogenesis, and newly-formed neuron survival in the hippocampus.23
Mechanism of action: a novel approach
to depression
Valdoxan has proven efficacy in animal models of
depression in which melatonin and 5HT2C receptor
antagonists have no effect. The preclinical studies
show that this efficacy is dependent on both melatonergic receptor agonism and 5HT2C receptor
Better quality remission in depression: Valdoxan, the first melatonergic antidepressant – Muñoz
VALDOXAN
antagonism. The results suggest that neither the
affinity for either receptor alone nor a simple combination of the two pharmacologic activities suffice
to provide antidepressant activity. The receptors
could therefore be acting in synergy (rather than
in combination) to achieve antidepressant efficacy
via complete regulation of circadian rhythms. Several lines of evidence substantiate this proposed
mode of action: (i) high MT1 , MT2 , and 5HT2C receptor density in the suprachiasmatic nucleus and
hippocampus24-26; (ii) involvement of all these receptors in the regulation of circadian rhythm27,28;
(iii) circadian expression of MT1 and 5HT2C receptors29,30; and (iv) beneficial effects on sleep by melatonergic agonists and slow-wave sleep promotion
by 5HT2C receptor antagonists.27,31 This mechanism
of action represents a true innovation in antidepressant therapy.
HAM-D total score
25
20
*
n=119
15
**
n=116
***
0
W0
W2
W4
W6
Figure 2. Mean Hamilton Rating Scale for Depression
(HAM-D) total scores (adjusted for center and baseline; last observation carried forward) in the Valdoxan
(25-50 mg) and placebo groups of a randomized, double-blind, parallel group trial. *P<0.05; **P<0.01;
***P<0.001.
After reference 33: Olié JP, Kasper S. Efficacy of Valdoxan, a MT1/MT2
receptor agonist with 5-HT2C antagonistic properties, in major depressive disorder. Int J Neuropsychopharmacol. 2007;10:661-673.
Copyright © 2007, CINP.
Clinical studies:
Valdoxan is a potent antidepressant,
providing faster, fuller, and
more sustained relief
ment with Valdoxan in CGI-S score, number of responders, and time to first response survival analysis.
Efficacy in severe depression
In all three placebo-controlled pivotal studies, Valdoxan showed significant efficacy in the severe subpopulation, irrespective of the severity criteria used
(baseline HAM-D score 25 or HAM-D score 25
and CGI score 5). Meta-analysis of the pooled
study data (296 patients randomized to Valdoxan
and 295 to placebo) showed that when the population was divided into subgroups using increasing
cut-off HAM-D scores (stepwise from HAM-D 24 to
HAM-D 30) at inclusion, antidepressant efficacy
persisted irrespective of baseline severity (Δ=2.06;
P=0.021 and Δ=4.45; P=0.025 for HAM-D cut-offs
of 22-25 and >30) (Figure 3).35
Three short-term pivotal studies have demonstrated antidepressant efficacy versus placebo, while
comprehensive evaluations have shown that this
efficacy is sustained across all grades of depression.
Efficacy has also been evaluated in head-to-head
comparisons versus venlafaxine and sertraline.
Superior efficacy to placebo: short-term studies
Dose-ranging study
This was an international, double-blind, randomized, phase 2, parallel-group efficacy study over 8
weeks in 711 patients with MDD (including bipolar II disorder) of Valdoxan 1-5 mg or 25 mg given in the evening versus placebo using paroxetine
20 mg as the internal validator.32 The efficacy end
point (mean decrease in total score on the 17-item
Hamilton Rating Scale for Depression [HAM-D17])
confirmed the efficacy of Valdoxan 25 mg versus
placebo (Δ 2.57; P=0.034), as did the secondary end
points: Montgomery-Åsberg Depression Rating
Scale (MADRS; P=0.016), Clinical Global Impression–Severity (CGI-S) scale (P=0.049), number of
responders (61.5% vs 46.3%; P=0.036), and time to
first response (P=0.008).
Flexible dose studies
Two international, double-blind, randomized, parallel-group studies confirmed the antidepressant
efficacy of Valdoxan 25 mg versus placebo over 6
weeks.33,34 The dose could be increased up to 50 mg
if improvement was insufficient (based on predetermined cut-offs on the HAM-D17 and CGI scales)
after 2 weeks. The design was unique insomuch as
investigators and patients were blinded to the criteria for the dose increase and to the increase itself,
which was performed via an interactive voice system.
In both studies (n=23833 and n=21234), Valdoxan was
more effective than placebo in terms of the mean
HAM-D score in both the total population (Δ=3.44;
P<0.001 [Figure 2] and Δ=2.3; P=0.026) and doseadjusted population (Δ=3.71; P=0.018 and Δ=3.13;
P=0.045). Secondary end points showed improve-
Placebo
Valdoxan 25-50 mg
Δ*= 3.44
30
Superior efficacy to placebo: long-term studies
A relapse prevention study randomized responders
to Valdoxan 25-50 mg after 8-10 weeks to continue
Total HAM-D at inclusion
2.06 (P=0.021)
[22-25] (n=233)
3.31 (P=0.003)
[26-27] (n=193)
3.46 (P<0.001)
[28-30] (n=215)
4.45 (P=0.025)
–4
–3
–2
–1
0
Favors placebo
1
2
3
4
5
[>30] (n=80)
6
7
8
9
Favors Valdoxan
Figure 3. Pooled results of positive placebo-controlled studies on Valdoxan showing
antidepressant efficacy in severely depressed patients. Hamilton Rating Scale for
Depression (HAM-D) difference from placebo (6-8 weeks; last observation carried
forward). The efficacy of Valdoxan 25-50 mg is maintained whatever the degree of
severity at inclusion.
After reference 35: Montgomery SA, Kasper S. Severe depression and antidepressants. Focus on a pooled
analysis of placebo-controlled studies on agomelatine. Int Clin Psychopharmacol. 2007;22:283-291.
Copyright © 2007, Lippincott Williams and Wilkins.
Better quality remission in depression: Valdoxan, the first melatonergic antidepressant – Muñoz
MEDICOGRAPHIA, VOL 31, No. 2, 2009 177
VALDOXAN
Valdoxan or placebo for 6 months (or an optional
10 months).36,37 The differences in cumulative relapse rates (21.7% vs 46.6% and 23.9% vs 50%) at
both 6 and 10 months, respectively, were highly significant: P<0.0001, with nearly 8 in 10 patients relapse-free on Valdoxan, and a risk of relapse half that
of placebo. The effect was confirmed in the severely depressed subpopulation.
pects of depression. Valdoxan also eliminates daytime drowsiness. Two studies have evaluated the effect of Valdoxan on sleep-wake patterns using objective measures (polysomnography) and subjective
questionnaires. Other information has come from
subanalysis of the HAM-D sleep items in the pivotal
studies.41
Objective studies
In the first study,42 depressed patients received Valdoxan 25 mg in the evening for 42 days under open
conditions with regular polysomnography. The results showed increased sleep efficiency (P=0.05),
decreased intrasleep awakenings (P=0.041), increased slow-wave sleep (stages 3 and 4) both absolutely (P=0.037) and relative to the total sleep
time (P=0.022), absence of change in total rapid eye
movement sleep, and gradual improvement in the
delta sleep ratio. Redistribution of slow-wave sleep
through the night normalized sleep architecture.
The head-to-head comparison versus sertraline
used actimetry to demonstrate improvement by the
end of week 1 on Valdoxan in the objective sleepwake parameters sleep efficiency, sleep latency, and
sleep-wake time.43
Subjective studies
A 6-week randomized double-blind study compared
the effect of Valdoxan 25-50 mg and venlafaxine 75150 mg on subjective sleep quality and integrity of
behavior using the Leeds Sleep Evaluation Questionnaire (LSEQ) and Visual Analog Scale (VAS).38
Efficacy versus venlafaxine and sertraline:
short-term and long-term studies
A head-to-head comparative study randomized patients to Valdoxan 25-50 mg (n=163) and venlafaxine 75-150 mg (n=165) in a flexible dose design for
6 weeks extending up to 6 months.38 Improvement
on the CGI-improvement (CGI-I) scale was much
greater as early as the first week in the Valdoxan
patients (Δ=0.39; 95% confidence interval [CI],
0.20-0.58; P<0.0001) and remained so at both 6
weeks (Δ=0.32; 95% CI, 0.06-0.58; P=0.016) and
6 months (Δ=0.32; 95% CI, 0.04-0.6; P=0.025).
Another head-to-head comparative study randomized patients to Valdoxan 25-50 mg (n=150)
and sertraline 50-100 mg (n=157) for 6 weeks up
to 6 months.39 The response rate was significantly
higher with Valdoxan at week 2 (20% vs 10.9%;
P=0.027), and at 6 weeks, the scores on the three
assessment scales were significantly superior —
HAM-D (Δ=1.68; P=0.031), CGI-I (Δ=0.29; P=0.023),
and CGI-S (Δ=0.28; P=0.043). At 6 months, the response rate was significantly higher with Valdoxan
(76% vs 63.5%; P=0.017).
Ease of getting to sleep
Anxiolytic activity
Better quality remission with Valdoxan
Underlying the antidepressant efficacy of Valdoxan
is a property not shared by any other available treatment: regulation of the sleep-wake rhythm, which
complements and enhances a number of equally
unique clinical benefits ensuring better quality remission. Sedation, daytime somnolence, sexual dysfunction, antidepressant discontinuation syndrome,
and weight gain are all remarkable by their absence
on Valdoxan therapy, contributing to excellent overall tolerability.
Regulation of the sleep-wake rhythm:
unique to Valdoxan
As expected from its pharmacological profile, Valdoxan restores the disturbed sleep-wake rhythm,
which is one of the earliest and most disabling as178 MEDICOGRAPHIA, VOL 31, No. 2, 2009
LSEQ score (mm) at week 1
P=0.007
60
55
50
VAS score (mm) at week 1
65
Improvement in
daytime alertness
65
Improvement in
ease of getting to sleep
Valdoxan is not only an effective antidepressant, it
also relieves the associated anxiety, as shown using the Hamilton Anxiety Scale (HAMA) in a doseranging study versus placebo (Δ=3.43; P=0.011),
and in the meta-analysis of positive placebo-controlled studies in which Valdoxan patients scored
significantly better versus placebo in the psychic
and somatic anxiety items (items 10 & 11) of the
HAM-D scale, including patients not on concomitant benzodiazepines.40 Valdoxan also outperformed
sertraline in terms of HAMA scores at 6 weeks
(Δ=2.36; 95% CI, 0.45-4.26; P=0.016).39
Daytime alertness
Venlafaxine
75-150 mg
Agomelatine
25-50 mg
P<0.001
60
55
50
45
Venlafaxine
75-150 mg
Agomelatine
25-50 mg
Figure 4. Effects of Valdoxan 25-50 mg and venlafaxine 75-150 mg after 1 week of
treatment on the first item in the Leeds Sleep Evaluation Questionnaire (LSEQ; “ease
of getting to sleep”; left panel) and on daytime functioning (Visual Analog Scale [VAS];
right panel). Valdoxan regulates sleep better than venlafaxine with improvement in
patients’ daytime condition from the first week of treatment. Based on data from
reference 38.
It showed significantly greater improvement with
Valdoxan in the LSEQ items “getting to sleep”
(P=0.007) and “quality of sleep” (P=0.015) in week 1.
This effect persisted until the end of treatment. Significantly greater improvement was also noted in
“daytime alertness” and “sensation of well-being”
(P0.001 in week 1), indicating earlier improvement of daytime functioning (Figure 4). The study
versus sertraline replicated these findings, showing
earlier improvement in “getting to sleep” (P<0.01)
and “quality of sleep” (P=0.025) with Valdoxan as
early as week 1.
Better quality remission in depression: Valdoxan, the first melatonergic antidepressant – Muñoz
VALDOXAN
Placebo n=402
30
Valdoxan 25/50 mg n=508
Patients (%)
25
20
Superior tolerability to conventional
15
antidepressants
Tolerated as well as placebo, including with
10
5
0
symptoms on the Discontinuation Emergent Signs
and Symptoms (DESS) checklist after 1 week. Abrupt
replacement of paroxetine by placebo, on the other
hand, induced many more emergent symptoms than
in patients remaining on paroxetine (P<0.001).46
[–9%; –7%]
[–5%; –3%]
[–1%; 1%]
[3%; 5%]
[7%; 9%]
Weight variation (%)
Valdoxan improves sleep through the night:
effect on early and middle insomnia and early
awakening
In the subanalysis of the three pivotal efficacy studies, Valdoxan 25-50 mg was significantly better than
placebo on HAM-D sleep subscales in all three phases of sleep: early insomnia (P<0.001), middle insomnia (P=0.015), and early awakening (P=0.006). The
difference versus placebo remained significant even
without the HAM-D sleep items.41
Figure 5. Body weight
variation compared
between Valdoxan and
placebo in the major depressive disorder (MDD)
safety sets of long-term
placebo-controlled trials.
Body weight variation
was similarly infrequent
in the Valdoxan and
placebo groups.
Preservation of sexual functioning
There has been no evidence at any time in its development that Valdoxan impairs sexual function.
Sexual emergent adverse events were few and similar on Valdoxan and placebo. A 12-week specific
study versus venlafaxine using the Sex Effects Scale
in 276 depressive patients, of whom 193 were sexually active at baseline, showed higher scores for
pre-orgasm and orgasm with Valdoxan 50 mg than
with venlafaxine 150 mg; the same was true in patients who achieved remission (n=111).44
A study in 92 healthy male volunteers randomized to Valdoxan (25 or 50 mg), paroxetine 20 mg,
or placebo for 8 weeks confirmed the nil-effect on
sexual function using the Psychotropic-Related
Sexual Dysfunction-Salamanca Sex Questionnaire
(PRSEX-DQ-SALSEX). Sexual dysfunction was
markedly less frequent (P<0.0001) in each Valdoxan group (25 mg, 22.7%; 50 mg, 4.8%) than in the
paroxetine group (85.7%). In the placebo group,
8.7% of volunteers reported sexual dysfunction. Volunteer reports of moderate or severe sexual dysfunction were 4.5% for Valdoxan 25 mg, 4.8% for
Valdoxan 50 mg, 61.9% for paroxetine 20 mg, and
0% in the placebo group (P<0.0001 for Valdoxan
versus paroxetine).45
Absence of the discontinuation syndrome
Abrupt discontinuation of conventional antidepressant treatment can trigger some very disturbing
symptoms that typically last several days and include nausea, delirium, chills, and sleep disturbance.
This does not occur with Valdoxan. A randomized,
double-blind and placebo-controlled study explored
the possibility of discontinuation syndrome with
Valdoxan versus paroxetine as the active control. In
stable MDD remitters, abrupt cessation of the 12week treatment with Valdoxan 25 mg induced no
respect to body weight
In the short-term studies, the percentages of patients reporting at least one treatment-emergent
adverse event (TEAE) on Valdoxan were low and
similar to placebo, as were the overall rates of TEAEs
and their nature. The most frequent adverse events
reported in both groups were headache, nausea, and
fatigue.47 No clinical relevant changes in body weight
(Figure 5) were observed on Valdoxan 25-50 mg.
Most TEAEs occurred in the first 2 weeks of treatment and were mild to moderate, transient, and
self-resolving. Rates of dropout due to adverse events
were similar for Valdoxan 25 mg (5.5%), Valdoxan
50 mg (5.2%), and placebo (5.2%).
Valdoxan has a cardiovascular safety profile similar to that of placebo. Hematology and clinical
chemistry show nil of note. There have been cases
of liver enzyme elevation (1.1%; without clinical
signs and reversible). Valdoxan has no apparent effect on hormone levels, including prolactin.
Superior tolerability to venlafaxine, paroxetine,
and sertraline
The safety profile of Valdoxan 25-50 mg compared
favorably with venlafaxine 75-150 mg over treatment for 6 or 12 weeks.38,44 Fewer patients withdrew
due to TEAEs on Valdoxan (4.2% vs 13.2% and 2.2%
vs 8.6% after 6 and 12 weeks). There were also fewer total TEAEs, in particular gastrointestinal disturbances and dizziness, on Valdoxan. An 8-week
dose-ranging study32 showed no difference in tolerability between Valdoxan 25 mg and placebo, whereas patients receiving paroxetine 20 mg reported significantly more gastrointestinal TEAEs than those
on placebo (P<0.05) (Table I). Comparison versus
sertraline 50-100 mg showed fewer TEAEs causPatients reporting emergent adverse events (%)
Headache
Anxiety
Abdominal pain
Diarrhea
Nausea
Somnolence
Depression
Insomnia
Valdoxan
(25 mg/day)
Placebo
Paroxetine
(20 mg/day)
6.6
3.6
3.6
3.6
2.9
2.9
2.9
2.9
8.6
3.6
3.6
2.9
4.3
5.0
2.7
2.9
8.2
2.7
3.4
4.1
17.0*
7.5
4.3
4.8
Table I. Main emergent adverse events (% patients in the safety set) reported by at least 3.5% of patients in any treatment group. Data from all
patients who received at least one dose of treatment (even without efficacy
evaluation). *P<0.05 (compared with placebo).
Modified from reference 32: Lôo H, Hale A, D’Haenen H. Determination of the dose of
agomelatine, a melatoninergic agonist and selective 5-HT2C antagonist, in the treatment of major depressive disorder: a placebo-controlled dose range study. Int Clin Psychopharmacol. 2002;17:239-247. Copyright © 2002, Lippincott Williams & Wilkins, Inc.
Better quality remission in depression: Valdoxan, the first melatonergic antidepressant – Muñoz
MEDICOGRAPHIA, VOL 31, No. 2, 2009 179
VALDOXAN
ing dropout after 6 weeks with Valdoxan 25-50 mg
(2.6% vs 11.3%).
The unique receptor profile accounts for the excellent tolerability of Valdoxan:
Because Valdoxan has no affinity for serotonin
or dopamine transporters, does not increase extracellular serotonin levels, and does not desensitize
5HT1A receptors (unlike TCAs/SSRIs/SNRIs), it does
not cause nausea, sexual dysfunction, central nervous system disorders, agitation, nervousness, or
insomnia.
Because Valdoxan has no affinity for muscarinic
receptors (unlike TCAs), it does not cause dry mouth,
somnolence, blurred vision, urinary retention, or
memory deficit.
Because Valdoxan has no affinity for histamine
receptors (unlike TCAs and mirtazapine), it does not
cause sedation or weight gain.
Because Valdoxan has no affinity for noradrenergic receptors or norepinephrine transporters (unlike TCAs, mirtazapine, and SNRIs), it has no cardiovascular effects and does not cause panic attacks or
anxiety.
Valdoxan: superior treatment adherence
Proven short-term and long-term efficacy combines
with these unique clinical benefits to generate better treatment adherence than with the active comparators used in the specific studies. Meta-analysis
of the two head-to-head studies showed that more
patients continued treatment with Valdoxan beyond 6 months than with venlafaxine and sertraline
(69.4% vs 61.5%; P<0.05).
REFERENCES
1. Moeller HJ. Outcomes of major depressive disorder:
the evolving concept of remission and its implications
for treatment. World J Biol Psychiatry.2008;9:102-114.
2. Kramer MS, Cutler N, Feighner J, et al. Distinct
mechanism for antidepressant activity by blockade of
central substance P receptors. Science. 1998;281:
1640-1645.
3. Griebel G, Simiand J, Steinberg R, et al. 4-(2-Chloro4-methoxy-5-methylphenyl)-N-[(1S)-2-cyclopropyl1-(3-fluoro-4-methylphenyl)ethyl]5-methyl-N-(2propynyl)-1, 3-thiazol-2-amine hydrochloride (SSR
125543A), a potent and selective corticotrophin-releasing factor(1) receptor antagonist. II. Characterization in rodent models of stress-related disorders.
J Pharmacol Exp Ther. 2002;301:333-345.
4. Souetre E, Salvati E, Wehr TA, et al. Twenty-fourhour profiles of body temperature and plasma TSH in
bipolar patients during depression and during remission and in normal control subjects. Am J Psychiatry.
1988;145:1133-1137.
5. Souetre E, Salvati E, Belugou JL, et al. Circadian
rhythms in depression and recovery: evidence for
blunted amplitude as the main chronobiological abnormality. Psychiatry Res. 1989;28:263-278.
6. Wirz-Justice A. Biological rhythms in mood disorders. In: Bloom FE, Kupfer DJ, eds. Psychopharmacology: The Fourth Generation of Progress. New York,
NY: Raven Press; 1995:999-1017.
7. Audinot V, Mailliet F, Lahaye-Brasseur C, et al. New
selective ligands of human cloned melatonin MT1 and
MT2 receptors. Naunyn-Shmiedeberg’s Arch Pharmacol. 2003;367:553-561.
8. Millan MJ, Gobert A, Lejeune F, et al. The novel
melatonin agonist Valdoxan (S20098) is an antagonist
at 5-hydroxytryptamine2C receptors, blockade of which
enhances the activity of frontocortical dopaminergic
and adrenergic pathways. J Pharmacol Exp Ther.
2003;306:954-964.
9. Hanoun N, Mocaër E, Boyer PA, et al. Differential
180 MEDICOGRAPHIA, VOL 31, No. 2, 2009
Conclusion
As the first melatonergic antidepressant, Valdoxan
is also the first new pharmacologic approach to
depression since the SSRIs. Its novel mode of action has been shown to answer most of the unmet
needs in depression therapy. By restoring biological rhythms, Valdoxan acts at the very core of depression, providing potent short- and long-term efficacy, exemplified in severely depressed patients,
and outperforming the standard therapeutic options in this regard, namely SSRIs and SNRIs. The
regulation of sleep-wake rhythms enables Valdoxan
to relieve the disturbed sleep experienced by depressed patients, and to do so uniquely—not only
without sedation or a hangover effect but with increased daytime alertness and preserved daily activities, all from the very first treatment week.
This novel mode of action also accounts for the
favorable side-effect profile of Valdoxan. The striking
absence of weight gain, sexual dysfunction, and discontinuation syndrome helps to secure unmatched
levels of treatment adherence.47
Conventional treatments achieve remission in
only a third of depressed patients. Even then the
remission is often of poor quality due to residual
symptoms. Its properties make Valdoxan unique in
the antidepressant armamentarium. It improves
quality of life during treatment and achieves better quality remission by acting directly on residual
symptoms. As such, Valdoxan may well represent
the antidepressant medication that patients and the
psychiatric community have been waiting for. effects of the novel antidepressant Valdoxan (S 20098)
versus fluoxetine on 5-HT1A receptors in the rat brain.
Neuropharmacology. 2004;47:515-526.
10. Millan MJ, Brocco M, Gobert A, et al. Anxiolytic
properties of Valdoxan, an antidepressant with melatoninergic and serotonergic properties: role of 5-HT2C
receptor blockade. Psychopharmacology. 2005;177:
1-12.
11. Papp M, Gruca P, Boyer PA, et al. Effect of Valdoxan in the chronic mild stress model of depression in
the rat. Neuropsychopharmacology. 2003;28:694-703.
12. Bertaina-Anglade V, Drieu la Rochelle C, Boyer
PA, et al. Antidepressant-like effects of agomelatine
(S 20098) in the learned helplessness model. Behav
Pharmacol. 2006;17:703-713.
13. Barden N, Shink E, Labbe M, et al. Antidepressant
action of Valdoxan (S 20098) in a transgenic mouse
model. Prog Neuropsychopharmacol Biol Psychiatry. 2005;29:908-916.
14. Bourin M, Mocaer E, Porsolt R. Antidepressantlike activity of S 20098 (Valdoxan) in the forced swimming test in rodents: involvement of melatonin and
serotonin receptors. J Psychiatry Neurosci. 2004;29:
126-133.
15. Van Reeth O, Weibel L, Olivares E, et al. Melatonin
or a melatonin agonist corrects age-related changes
in circadian response to environmental stimulus. Am
J Physiol Regul Integr Comp Physiol. 2001;280:
R1582-R1591.
16. Armstrong SM, McNulty OM, Guardiola-Lemaitre
B, et al. Successful use of S20098 and melatonin in
an animal model of delayed sleep-phase syndrome
(DSPS). Pharmacol Biochem Behav.1993;46:45-49.
17. Redman JR, Guardiola-Lemaitre B, Brown M, et
al. Dose dependent effects of S-20098, a melatonin agonist, on direction of re-entrainment of rat circadian
activity rhythms. Psychopharmacology. 1995;118:
385-390.
18. Grassi-Zucconi G, Semprevivo M, Mocaër E, et al.
Melatonin and its new agonist S 20098 restore synchronized sleep fragmented by experimental trypanosome infection in the rat. Brain Research Bulletin.
1996;39:63-68.
19. Corbach S, Schmelting B, Fuchs E, et al. Comparison of agomelatine and melatonin for effects in
chronically stressed tree shrews, an animal model of
depression. Eur Neuropsychopharmacol. 2007;7(suppl 4):S364.
20. Fuchs E, Corbach-Söhle S, Schmelting B, et al.
Effects of agomelatine compared to melatonin in psychosocially stressed tree shrews. Int J Neuropsychopharmacol. 2008;11(suppl 1):121.
21. Leproult R, Van Onderbergen A, L’Hermitte-Balériaux M, et al. Phase-shifts of 24-h rhythms of hormonal release and body temperature following early
evening administration of the melatonin agonist agomelatine in healthy older men. Clin Endocrinol. 2005;
63:298-304.
22. Papp M, Litwa E, Gruca P, et al. Anxiolytic-like activity of Valdoxan and melatonin in three animal models of anxiety. Behav Pharmacol. 2006;17:9-18.
23. Banasr M, Soumier A, Hery M, et al. Agomelatine, a new antidepressant, induces regional changes
in hippocampal neurogenesis. Biol Psychiatry. 2006;
59:1087-1096.
24. Boess FG, Martin IL. Molecular biology of 5-HT
receptors. Neuropharmacology. 1994;33:275-317.
25. Moyer RW, Kennaway DJ. Immunohistochemical
localization of serotonin receptors in the rat suprachiasmatic nucleus. Neurosci Lett. 1999;271:147-150.
26. Larson J, Jessen RE, Uz T, et al. Impaired hippocampal long-term potentiation in melatonin MT2 receptor-deficient mice. Neurosci Lett. 2006;393:23-26.
27. Dugovic C, Leysen JE, Wauquier A. Melatonin
modulates the sensitivity of 5-hydroxytryptamine-2
receptor-mediated sleep-wakefulness regulation in the
rat. Neurosci Lett. 1989;104:320-325.
28. Kennaway DJ, Moyer RW. Serotonin 5-HT2C ag-
Better quality remission in depression: Valdoxan, the first melatonergic antidepressant – Muñoz
VALDOXAN
onists mimic the effect of light pulses on circadian
rhythms. Brain Res. 1998;806:257-270.
29. Holmes MC, French KL, Seckl JR. Dysregulation
of diurnal rhythms of serotonin 5HT2C and corticosteroid receptor gene expression in the hippocampus
with food restriction and glucocorticoids. J Neurosci.
1997;17:4056-4065.
30. Masana MI, Benloucif S, Dubocovich ML. Circadian rhythm of MT1 melatonin receptor expression, in
the suprachiasmatic nucleus of the C3H/HeN mouse.
J Pineal Res. 2000;28:185-192.
31. Smith MI, Piper DC, Duxon MS, et al. Effect of SB243213, a selective 5-HT2C receptor antagonist, on the
rat sleep profile: a comparison to paroxetine. Pharmacol Biochem Behav. 2002;71:599-605.
32. Lôo H, Hale A, D’Haenen H. Determination of the
dose of agomelatine, a melatoninergic agonist and selective 5-HT2C antagonist, in the treatment of major
depressive disorder: a placebo-controlled dose range
study. Int Clin Psychopharmacol. 2002;17:239-247.
33. Olié JP, Kasper S. Efficacy of Valdoxan, a MT1/
MT2 receptor agonist with 5-HT2C antagonistic properties, in major depressive disorder. Int J Neuropsychopharmacol. 2007;10:661-673.
34. Kennedy SH, Emsley R. Placebo-controlled trial
of Valdoxan in the treatment of major depressive disorder. Eur Neuropsychopharmacol. 2006;16:93-100.
35. Montgomery SA, Kasper S. Severe depression and
antidepressants. Focus on a pooled analysis of place-
bo-controlled studies on agomelatine. Int Clin Psychopharmacol. 2007;22:283-291.
36. Goodwin GM, Rouillon F, Emsley R. Long term efficacy of agomelatine, a novel antidepressant, in the
prevention of relapse in out-patients with major depressive disorder. Eur Neuropsychopharmacol. 2007;
17(suppl 4):361.
37. Goodwin GM, Rouillon F, Emsley R. Long-term
treatment with agomelatine: prevention of relapse
in patients with major depressive disorder over 10
months. Eur Neuropsychopharmacol. 2008;18(suppl 4):S338.
38. Lemoine P, Guilleminault C, Alvarez E. Improvement in subjective sleep in major depressive disorder
with a novel antidepressant, agomelatine: randomized, double-blind comparison with venlafaxine. J Clin
Psychiatry. 2007;68:1723-1732.
39. Kasper S, Laigle L, Baylé F. Superior antidepressant efficacy of agomelatine versus sertraline: a randomised, double-blind study. Eur Neuropsychopharmacol. 2008;18(suppl 4):336.
40. Kennedy S, Lôo H, Olié JP. The antidepressant
efficacy of agomelatine is independent of gender, age,
body mass index and use of concomitant anxiolytics:
combined analysis of three placebo-controlled studies.
Int J Neuropsychopharmacol. 2008;11(suppl 1):193.
41. Guilleminault C, Quera-Salva MA. Impact of the
melatonergic antidepressant Valdoxan on sleep-wake
rhythms of depressed patients. Eur Neuropsycho-
pharmacol. 2006;16(suppl 4):317.
42. Quera Salva MA, Vanier B, Laredo J, et al. Major
depressive disorder, sleep EEG and agomelatine: an
open-label study. Int J Neuropsychopharmacol. 2007;
10:691-696.
43. Kasper S. Effect of agomelatine on rest-activity
cycle in patients with major depressive disorder compared to sertraline. Int J Neuropsychopharmacol.
2008;11(suppl 1):193.
44. Kennedy SH, Rizvi S, Fulton K, et al. A doubleblind comparison of sexual functioning, antidepressant efficacy, and tolerability between agomelatine
and venlafaxine XR. J Clin Psychopharmacol. 2008;
28:329-333.
45. Montejo AL, Prieto N, Terleira A, et al. Better sexual acceptability of agomelatine (25 and 50 mg) compared to paroxetine (20 mg) in healthy male volunteers. An 8-week, placebo controlled study using the
PRSEXDQ-SALSEX scale. J Psychopharmacol.Sept 18
2008. Epub ahead of print.
46. Montgomery SA, Kennedy SH, Burrows GD, et al.
Absence of discontinuation symptoms with Valdoxan
and occurrence of discontinuation symptoms with
paroxetine: a randomized, double-blind, placebo-controlled discontinuation study. Int Clin Psychopharmacol. 2004;19:271-280.
47. Rouillon F. Efficacy/tolerance profile of Valdoxan
and practical use in depressed patients. Eur Neuropsychopharmacol. 2005;15:S660-S661.
AMÉLIORER LA QUALITÉ DE LA RÉMISSION DANS LA DÉPRESSION :
VALDOXAN, LE PREMIER ANTIDÉPRESSEUR MÉLATONINERGIQUE
e fait que les antidépresseurs classiques confèrent rarement une rémission de bonne qualité suggère que d’autres
modèles que le système monoaminergique pourraient ouvrir la voie à un traitement efficace. La régulation des rythmes
circadiens en est l’illustration, fondée sur la perturbation de ces
rythmes chez les patients déprimés et leur l’importance dans l’évolution des l’épisodes dépressifs. Valdoxan (agomélatine) est le
premier antidépresseur mélatoninergique. Son profil d’action au
niveau des récepteurs lui confère une efficacité unique dans la régulation des troubles des rythmes circadiens. Valdoxan, en effet,
est le premier antidépresseur agoniste des récepteurs mélatoninergiques MT1 et MT2 et antagoniste des récepteurs sérotoninergiques 5HT2C . Son efficacité antidépressive a été démontrée
L
à court et long termes contre placebo, quelle que soit la sévérité
de la maladie, et s’est avérée supérieure à celle de la venlafaxine
et la sertraline aux différentes échelles d’évaluation, particulièrement celles utilisées en pratique clinique. Cette efficacité
s’associe à des bénéfices cliniques singuliers tels que la régulation non sédative du cycle veille-sommeil et l’amélioration du
fonctionnement diurne, dès la première semaine de traitement.
Valdoxan n’altère pas les fonctions sexuelles ni la libido, il n’induit pas de syndrome de manque à l’arrêt du traitement ou de
prise de poids, et il est bien toléré, ce qui en fait un traitement
réellement innovant de la dépression, permettant un soulagement précoce des symptômes ainsi qu’une rémission complète
et prolongée.
Better quality remission in depression: Valdoxan, the first melatonergic antidepressant – Muñoz
MEDICOGRAPHIA, VOL 31, No. 2, 2009 181
I
N T E R V I E W
CLINICAL MANAGEMENT OF PATIENTS
FOLLOWING REMISSION FROM DEPRESSION
I n t e r v i e w w i t h W. C h o u c h a a n d
J . - F. A l l i l a i r e , F r a n c e
Left. Jean-François ALLILAIRE, MD, PhD
Right. Walid CHOUCHA, MD
CHU Pitié Salpêtrière, Service de Psychiatrie Adulte
Paris, FRANCE
For how long should an antidepressant
be prescribed following achievement of
remission, and at what dosage?
ontinuation or consolidation
therapy is systematically indicated once a patient has responded to
acute antidepressant treatment, whatever
the severity, nature, or number of depressive episodes. However, recommendations
for the length of this therapy vary widely
from 4 to 12 months, according to different authors. Some consider 4 to 9
months of continuation antidepressant
C
A
treatment to be sufficient. Others recommend approximately 6 months of continuation therapy if there are residual
symptoms, and 4 months if there is complete remission with no residual symptoms. More recently, prudent authors
have recommended 9 to 12 months of
continuation therapy whether residual
symptoms are present or not.1 During
this phase, the antidepressant should be
continued at the same dose that induced
remission. All studies have shown considerable benefit from continuation therapy,
with relapse rates at least halved. Continuation psychotherapy can be added
to continuation medication following an
acute phase response, whether to a single medication or a combination.
In certain situations, there is no need
for continuation antidepressant treatment to be followed by maintenance therapy: (i) patients presenting with a first
nonsevere depressive episode and aged
<55 years old (for some authors <50 years
old and for others <60 years old). Some
authors recommend continuation treatment for 4 to 6 months in this group of
continuation phase of antidepressant treatment is systematically indicated once a patient has responded to
acute-phase antidepressant treatment, and this should be
followed by a maintenance treatment phase in certain clinical
situations. The length of such phases remains controversial. However, the antidepressant dose during the continuation and maintenance phases should be the same as that which induced remission. Full remission means that the patient no longer meets the
criteria for a depressive disorder and has minimal symptoms at
most during 2 months or more. We generally consider that full
remission has occurred when the Hamilton Rating Scale for Depression score is less than 8. Consequently, residual symptoms
may persist even after the patient has achieved full remission,
and they are considered as an important risk factor for relapse
and long-term social dysfunction. The prevalence of residual
symptoms in remitted patients is estimated to be 30% to 50%.
They mainly include insomnia, anxiety, cognitive disturbances,
apathy, and fatigue/somnolence. Management of these symptoms
includes optimizing the antidepressant dose and duration, assess182 MEDICOGRAPHIA, VOL 31, No. 2, 2009
patients; and (ii) patients presenting with
a second nonsevere depressive episode
after >3 years of remission (for some authors, more than 5 years). Some authors
recommend continuation treatment for
6 to 12 months in this group of patients.
There is a 30% to 80% chance that these
two groups of patients will not show relapse or recurrence during 5 years of
follow-up after their last depressive exacerbation. After a full continuation phase,
antidepressant treatment discontinuation should then be gradual, and if there
is a return of the depressive symptoms,
SELECTED
ABBREVIATIONS AND ACRONYMS
BDI
DSM
Beck Depression Inventory
Diagnostic and Statistical Manual of Mental Disorders
HAM-D Hamilton Rating Scale for
Depression
MADRS Montgomery–Åsberg Depression
Rating Scale
PHQ-9 9-item Patient Health Questionnaire
ing and enhancing adherence to treatment, adding psychotherapies like interpersonal or cognitive therapy (with better evidence
for the latter), and augmenting current antidepressant treatment
with adjunctive pharmacotherapy. Other important risk factors
for relapse may include comorbid personality, somatic or social
disorders, and recurrent depression. Lifestyle recommendations
for maintaining remission may include regular physical exercise,
more balanced dietary habits, more healthy sleep, as well as better planning and organization of different daily life activities.
Medicographia. 2009;31:182-185.
(see French abstract on page 185)
Keywords: depression; continuation therapy; maintenance
therapy; remission; relapse; recurrence; residual symptom;
adjunctive psychotherapy; adjunctive pharmacotherapy;
lifestyle recommendation
www.medicographia.com
Address for correspondence: Dr Walid Choucha, CHU Pitié Salpêtrière,
Service de Psychiatrie Adulte, 47, boulevard de l’Hôpital, 75013 Paris, France
(e-mail: [email protected])
Clinical management of patients following remission from depression – Choucha and Allilaire
INTERVIEW
one should revert to the effective dose
with a further attempt at withdrawal after at least a further 4 to 6 months of
continuation treatment (a further 9-12
months according to some authors). Clinical experience indicates that this phenomenon usually reflects impending relapse. If such patients relapse again
during a later drug withdrawal trial, a
period of maintenance treatment then
becomes appropriate.
Maintenance, prophylactic, or preventive therapy is either of lifetime duration
or for at least 5 years.2 Lifetime maintenance treatment is indicated in the following situations: (i) patients presenting
with three or more depressive episodes;
(ii) patients presenting with their first
depressive episode over the age of 55
years (for some authors this is >60 years
of age); and (iii) patients presenting with
a second depressive episode when over
the age of 40 years.
In the following situations, treatment
should be maintained for at least 5 years
(3 years for some authors and 4 years
for others): (i) patients presenting with
a second depressive episode while aged
less than 40 years; (ii) patients presenting with a second depressive episode after
less than 3 years (for some authors <5
years) of maintained remission; (iii) patients presenting with a second depressive
episode after more than 3 years of remission (for some authors >5 years) but experiencing a severe episode (presence of
marked suicidal thoughts/actions, psychotic, melancholic or catatonic features,
etc); and (iv) patients presenting with a
severe first depressive episode. A total of
70% to 90% of these 4 groups of patients
are at risk of relapse or recurrence within 5 years if medication is stopped.
In all cases of maintenance therapy, the
antidepressant should be maintained at
the same dosage that induced remission.3
What results should a clinician
expect in terms of remission?
omplete or full remission4
means that the patient no longer meets the criteria for a depressive
disorder and has minimal symptoms at
most during 2 months or more. We generally consider that full remission has
occurred either when the Hamilton Rating Scale for Depression (HAM-D) score
is less than 8, the 9-item Patient Health
Questionnaire (PHQ-9) score is less than
5, or the Beck Depression Inventory (BDI)
score is less than 9.
The three items most frequently judged
by patients to be very important in determining remission are the presence of
C
features of positive mental health such
as optimism and self-confidence, a return
to one’s usual, normal self, and a return
to usual levels of functioning.
A patient is considered to be in recovery when remission is maintained for
beyond 6 months. The term partial remission means either that the patient
no longer meets the criteria for a depressive disorder and has minimal symptoms
at most for less than 2 months, or has
more than just minimal symptoms for 2
months or more. We generally consider
that partial remission has occurred when
the HAM-D score is over 7 (between 8
and 17 or 18).
Response means that there is a robust
improvement in depressive symptoms
with treatment (a reduction of 50% or
more in HAM-D or Montgomery–Åsberg
Depression Rating Scale [MADRS] scores).
Partial response means an improvement in depressive symptoms with treatment, but without reaching response
(only 25%-49% reduction in symptoms).
Nonresponse means a relative lack of
symptom improvement (less than 25%
reduction in symptoms). As we can see,
residual symptoms may persist even after
the patient has achieved full remission,
as defined by even the most conservative
criteria. Their prevalence in remitted
patients is estimated to be 30% to 50%.
Relapse occurs when depressive symptoms fulfilling the DSM (Diagnostic and
Statistical Manual of Mental Disorders)
criteria for a major depressive disorder
arise during either partial or full remission. Recurrence occurs when depressive
symptoms fulfilling the DSM criteria for
a major depressive disorder arise during
the period of recovery. Rebound means
a return of the original depressive symptoms but with greater intensity, and withdrawal refers to the development of different symptoms that are related to the
stopping of a drug treatment.
When is adjunctive therapy such as
psychotherapy needed?
he two main recommended
adjunctive psychotherapies in
depression are interpersonal therapy and
cognitive therapy. Their indication in
managing residual symptoms and preventing relapse and recurrence is, however, less well established. Trials of interpersonal therapy for the prevention of
recurrence have shown some benefits,
but the effects are weaker than those of
drugs, and any additional benefit in combination with drugs is limited.
There is better evidence for the effects
of cognitive therapy in preventing relapse
T
Clinical management of patients following remission from depression – Choucha and Allilaire
and recurrence, and there is an emerging indication for its addition to antidepressants, particularly when residual
symptoms are present. One randomized,
controlled study of cognitive therapy 5
involved 158 patients with recent major
depression who were partially remitted
with antidepressant treatment but who
had residual symptoms of 2 to 18 months’
duration. Patients randomized to continue antidepressant therapy with the addition of cognitive therapy (16 sessions
during 20 weeks with 2 subsequent booster sessions) showed significantly reduced
relapse rates at 68 weeks compared with
those who continued pharmacotherapy
alone (29% vs 47%).
Although the cognitive therapy group
showed a more significant increase in
remission rates at 20 weeks, the increase
was comparatively small and not reflected in the mean symptom ratings, particularly on ratings of worthlessness and
hopelessness.
It seems that the preventive effects of
cognitive therapy on relapse are more
powerful than the immediate effects on
residual symptoms. In a longitudinal
study,6 a total of 40 subjects with residual symptoms were randomized either to
modified cognitive therapy targeting anxiety and irritability, or to clinical management. Modified cognitive therapy
significantly reduced residual symptoms,
as well as relapse and recurrence rates
at 4 and 6 years, compared with the clinical management group (35% vs 70%).
In a subsequent study of recurrent depression,7 a total of 40 patients were randomized either to a therapeutic strategy
that included cognitive therapy, lifestyle
modification, and well-being therapy, or
to a control group. Modified cognitive
therapy significantly reduced recurrence
from 80% to 20% during 2 years.
What kinds of lifestyle recommendations are helpful for maintaining
remission in patients?
he first recommendation is
regular physical exercise. When
prescribing exercise, several caveats apply:
Anticipate barriers, as hopelessness
and fatigue can make physical exercise
difficult.
Keep expectations realistic, as some
patients are vulnerable to guilt and
self-blame if they fail to carry out the
regimen.
Introduce a feasible plan: walking,
alone or in a group, is often a good option. Paying attention while walking to
colors, smells, shapes, sounds, and people may be ideal.
T
MEDICOGRAPHIA, VOL 31, No. 2, 2009 183
INTERVIEW
Accentuate the pleasurable aspects of
exercise: the specific choice of exercise
should be guided by the patient’s preferences, and must be pleasurable.
A goal of 30 minutes of moderate-intensity aerobic exercise, 3-5 times a week
is recommended for otherwise healthy
adults.
Encourage adherence, as greater antidepressant effects are seen when training
continues beyond 16 weeks.
Relaxation exercises, whenever possible, are recommended, including meditation and even yoga!
Another lifestyle recommendation concerns dietary habits. Less of a caloric and
more of a balanced diet is recommended. It is better to have more meals rather
than bigger meals.
Recommendations regarding sleep
habits are also important. Activities including sport, television, and even reading should ideally be avoided before going to sleep. A steady sleep routine that
involves always trying to go to bed early
and at the same time everyday is recommended. The sleeping room should be
as uncluttered as possible, with no office
or work equipment in it, and should be
dedicated only for sleeping. The bed mattress for sleeping should be comfortable.
Siestas, if possible, may be very beneficial.
Finally, daily life activities should be
planned in such a way as to minimize
or avoid “hurrying.” Multiple activities
should not be performed simultaneously.
Rest after every activity is recommended. As residual symptoms affect those
parts of the brain responsible for high
concentration activities, planning, and
organization, patients should therefore
try to avoid activities that demand too
much of this, such as reading, writing,
the Internet, or watching the news, and
encourage activities like gardening, painting, or bird-watching.
What therapeutic approach should
be used for residual symptoms?
e should first try to address
treatment-emergent side effects. Indeed, significant overlap exists
between the residual symptoms of a depressive episode and the side effects of
antidepressants. A systematic assessment
of symptoms prior to beginning pharmacological treatment can help to distinguish between residual symptoms and
treatment side effects. If dose optimization improves a patient’s symptoms, one
may assume that they are likely to have
been residual. On the other hand, if op-
W
184 MEDICOGRAPHIA, VOL 31, No. 2, 2009
timization has no effect or worsens symptoms, clinicians may assume that these
symptoms probably represent side effects
of pharmacologic therapy, as the intensity of the side effects is typically dosedependent. Second, we should diagnose
and treat any comorbid medical or psychiatric conditions, such as substance
abuse, since these conditions may cloud
the assessment of symptoms, whether
related to the underlying depressive disorder or antidepressant treatment.
If it is decided that symptoms are likely
to be residual, clinicians may want to
optimize the antidepressant dose and
duration, assess and enhance adherence
to treatment, add psychotherapy (cognitive or interpersonal therapy), and augment current therapy with adjunctive
pharmacological treatment. By contrast,
if symptoms are likely to be side effects
of pharmacological treatment, clinicians
may want to decrease the antidepressant
doses or switch the patient to another
antidepressant. Adjunctive treatment,
however, may improve such symptoms
regardless of the underlying etiology. Such
adjunctive treatment includes the following8: (i) for anxiety, either a benzodiazepine (like lorazepam, alprazolam, or
clonazepam), buspirone, gabapentin, or
even an antipsychotic (olanzapine, quetiapine); (ii) for insomnia, we can add
either an hypnotic (like zolpidem or zopiclone), or lorazepam, or sedative antidepressants (like mirtazapine, mianserine,
or trazodone); (iii) for fatigue and somnolence, we can add either methylphenidate, modafinil, bupropion, or reboxetine; (iv) for apathy, we can also add either
methylphenidate, modafinil, bupropion,
or reboxetine; and (v) for cognitive disturbances, we can add either anticholinesterase medications (like donepezil),
methylphenidate, modafinil, bupropion,
reboxetine, or memantine.
atients at risk of relapse are
those who present with:
(i) three or more depressive episodes;
(ii) a second depressive episode occurring after the age of 40 years or one that
is associated with severe symptoms, or
one that occurs less than 3 years after
remission from their first episode (for
some authors, less than 5 years); (iii) a
first depressive episode occurring after
the age of 50 to 60 years (or before 20
years of age according to certain authors); (iv) a longer depressive episode;
(v) residual symptoms at remission10:
their presence appears to affect early
short-term relapse rather than overall
long-term risk of recurrence or number
of recurrences, and to be responsible for
impaired long-term social functioning.
There is some conflict in the literature
concerning factors like initial severity of
depression, life stressors, and personality
as predictors of residual symptoms. In
addition, residual symptoms are prevalent both in patients who have received
psychotherapy, as well as those treated
with pharmacotherapy; (vi) relapse at
medication withdrawal11; (vii) family
history of bipolar disorder or recurrent
depression; (viii) associated disorders on
axis 1 or 2; and (ix) bad social adjustment,
persistent stressful life events, or absence
of social support. REFERENCES
1. Reimherr FW, Amsterdam JD, Quitkin FM, et al.
Optimal length of continuation therapy in depression: a prospective assessment during long-term fluoxetine treatment. Am J Psychiatry.1998;155:12471253.
2. Kupfer DJ, Frank E, Perel JM, et al. Five-year outcome for maintenance therapies in recurrent depression. Arch Gen Psychiatry. 1992;49:769-773.
3. Franck E, Kupfer DJ, Perel JM, et al. Comparison
of full-dose versus half-dose pharmacotherapy in the
maintenance treatment of recurrent depression. J Affect Dis. 1993;27:139-145.
4. Frank E, Prien RE, Jarrett RB, et al. Conceptualization and rationale for consensus definitions of terms
in major depressive disorder. Remission, recovery, relapse and recurrence. Arch Gen Psychiatry.1991;48:
287-297.
5. Paykel E, Scott J, Teasdale J, et al. Prevention of relapse in residual depression by cognitive therapy. Arch
Gen Psychiatry. 1999;56:829-835.
6. Fava GA, Grandi S, Rafanelli C, Canestrari R, Morphy MA. Six-year outcome for cognitive behavioral
treatment of residual symptoms in major depression.
Am J Psychiatry. 1998;155:1443-1445.
7. Fava GA, Rafanelli C, Grandi S, Conti S, Belluardo
P. Prevention of recurrent depression with cognitive
behavioral therapy: preliminary findings. Arch Gen
Psychiatry. 1998;55:816-820.
8. Fava M. Pharmacological approaches to the treatment of residual symptoms. Journal of Psychopharmacology. 2006;20:29-34.
9. Stahl SM. Essential Psychopharmacology. 2nd
ed. New York, NY: Cambridge University Press; 2000.
10. Kennedy N, Paykel ES. Residual symptoms at remission from depression: impact on long-term outcome. J Affect Dis. 2004;80:135-144.
11. Paykel ES. Continuation and maintenance therapy in depression. Br Med Bull. 2001;157:145-159.
It seems that the risk and severity of
residual symptoms is significantly less
with serotonin and norepinephrine reuptake inhibitors than with selective serotonin reuptake inhibitors9; the ideal
scenario would be an antidepressant that
addressed residual symptoms at the same
time as other symptoms, thus removing
the need for adjunctive therapy.
When should the clinician and the
patient suspect a relapse?
P
Clinical management of patients following remission from depression – Choucha and Allilaire
INTERVIEW
PRISE
EN CHARGE CLINIQUE DES PATIENTS APRÈS LA RÉMISSION D’UNE DÉPRESSION
orsqu’un patient a répondu à un traitement antidépresseur d’attaque, une phase de consolidation est systématiquement indiquée et doit, dans certaines situations cliniques, être poursuivie par une phase d’entretien. Si la durée de
ces différentes phases ne fait pas l’objet d’un consensus, la posologie du traitement antidépresseur, quant à elle, doit être la
même pendant les phases de consolidation et d’entretien que celle
qui a permis la rémission. Une rémission complète signifie que le
patient ne remplit plus les critères d’un trouble dépressif et ne présente que des symptômes minimaux, pendant 2 mois ou plus ;
elle correspond généralement à un score inférieur à 8 à l’échelle
HDRS (Hamilton Rating Scale for Depression). Les symptômes
résiduels, qui peuvent donc persister même après la rémission
complète, sont considérés comme un facteur de risque important
de rechutes et de handicap social à long terme. La prévalence des
L
symptômes résiduels chez les patients en rémission est estimée
entre 30 et 50 %. On y trouve principalement l’insomnie, l’anxiété, les troubles cognitifs, l’apathie, la fatigue et la somnolence.
Leur prise en charge comporte l’optimisation de la dose et de la
durée du traitement antidépresseur, l’évaluation et l’amélioration
de l’observance du traitement, l’ajout de psychothérapies interpersonnelles ou cognitives (l’efficacité de ces dernières étant mieux
prouvée) et l’association au traitement antidépresseur en cours
d’un traitement pharmacologique adjuvant. La personnalité comorbide, les troubles somatiques et sociaux et la dépression récurrente sont également des facteurs de risque important de rechute.
Pour contribuer au maintien de la rémission, on peut recommander un exercice physique régulier, de meilleures habitudes alimentaires, un sommeil plus équilibré ainsi qu’une meilleure planification et organisation des activités de la vie quotidienne.
Clinical management of patients following remission from depression – Choucha and Allilaire
MEDICOGRAPHIA, VOL 31, No. 2, 2009 185
F
O C U S
INCREASING THE LEVEL OF REMISSION
IN DEPRESSION BY THE SEQUENTIAL USE OF
PHARMACOTHERAPY AND PSYCHOTHERAPY
by G. A. Fava and D. Visani, Italy
everal investigations have suggested the usefulness of a sequential way of integrating pharmacotherapy and psychotherapy in depression.1 Administration of treatments in sequential
order is a common practice in clinical medicine,
particularly when treatment fails. This sequential
administration of treatments also occurs in clinical
psychiatry. It may involve switches to different types
of drugs, as is often the case in drug-refractory depression, an increase or decrease in dose, augmentation, or change to a different drug.2 There are also
examples of changes of types of treatment: use of
antidepressants after unsuccessful cognitive behavioral therapy (CBT) in depression,3 and of CBT in
the management of drug-resistant major depressive
illness.4
The recent findings of the largest depression trial, Sequenced Treatment Alternatives to Relieve Depression (STAR*D), have provided a dramatic illustration of the difficulties in achieving recovery from
depressive illness by using pharmacological strategies.5 The aim of the trial was to apply the best pharmacological strategies to obtain remission in major
depression. A sample of 3671 patients was treated
with citalopram in an open fashion: only 36.8% of
patients remitted. Those who did not recover were
submitted to four sequential steps involving switching, augmentation, and combination strategies,
S
T
Giovanni A. FAVA,*† MD
Dalila VISANI,* PsyD
* Department of Psychology
University of Bologna
Bologna, ITALY
† Department of Psychiatry
State University of
New York at Buffalo
Buffalo, NY, USA
here is increasing literature on the bleak long-term outcome of depression in terms of relapse and recurrence. This
unsatisfactory outcome seems to be associated with the
presence of substantial residual symptomatology. If residual
symptoms are the rule after completion of drug or psychotherapeutic treatment, and their presence has been correlated with poor
outcome — as residual symptoms upon recovery may progress
to become prodromal symptoms of relapse—then treatment directed toward residual symptoms may yield long-term benefits.
Treatments that are aimed at potentially different symptoms,
such as pharmacotherapy and psychotherapy, may thus be used
in a sequential order. One type of treatment (eg, psychotherapy)
may be employed to improve symptoms that the other type of
treatment (eg, pharmacotherapy) was unable to affect. Several
studies substantiate the clinical advantages of the sequential use
of psychotherapy after pharmacological treatment. There has been
little research on other forms of sequential treatment in depres186 MEDICOGRAPHIA, VOL 31, No. 2, 2009
based on available literature. The recovery rate was
low and difficult to attribute to specific effects of
citalopram, since there was a variety of nonspecific
therapeutic ingredients, as in other major trials.5-8
Because of the type of randomization that was chosen (equipoise-stratified randomization strategy),
the role of cognitive therapy could not be established, since the patients who opted for it were too
few (less than one third of participants).9 The STAR*D
results were rather disappointing. The cumulative rate of remission after 4 sequential steps was
67%.5 However, when sustained recovery was considered (taking into account relapse rates while on
treatment), the cumulative rate was 43%.10 This
means that the strenuous efforts after step one
(open treatment with citalopram) yielded an additional 6% of sustained recovery.11
In clinical medicine, there is also, however, another type of sequential treatment, which is not related to the partial remission or failure associated
with a specific therapy (sequence is performed regardless of the outcome of the first component, as a
preplanned strategy). This type of approach is based
on the awareness that one course of treatment is
unlikely to yield full disappearance of symptomatology. A related assessment strategy is the staging method, whereby a disorder is characterized according to seriousness, extension, and features.1,12,13
sion. It has also been suggested that the most effective drugs in
treating acute depression may not be the most suitable for postacute or continuation treatment. The sequential treatment of depressive illness does not fall within the realm of maintenance
strategies. It is an intensive two-stage approach, which is based on
the fact that one course of treatment with a specific tool (whether
pharmacotherapy or psychotherapy) is unlikely to hold a solution
to the complex array of symptoms in patients with depression.
The sequential model entails considerable implications for assessment and treatment planning.
Medicographia. 2009;31:186-191.
(see French abstract on page 191)
Keywords: depression; recovery; staging; sequential model;
cognitive behavioral therapy; wellbeing therapy
www.medicographia.com
Address for correspondence: Professor Giovanni A. Fava, Department of Psychology,
University of Bologna, Viale Berti Pichat 5, 40127 Bologna, Italy
(e-mail: [email protected])
Increasing remission levels with sequential pharmacotherapy and psychotherapy – Fava and Visani
FOCUS
SELECTED
ACT
CBT
DSM-IV
ETAU
IPT
MBCT
STAR*D
TAU
ABBREVIATIONS AND ACRONYMS
acceptance and commitment therapy
cognitive behavioral therapy
Diagnostic and Statistical Manual of
Mental Disorders–Fourth Edition
enhanced treatment as usual
interpersonal psychotherapy
mindfulness-based cognitive therapy
Sequenced Treatment Alternatives to
Relieve Depression (study)
treatment as usual
ing the sequential model in affective disorders is
the increasing awareness of the role of comorbidity.23,24 In major depression, two thirds of patients
meet the criteria for another axis I disorder (particularly anxiety disorders), and one third has two
or more disorders.25 It is thus unlikely that monotherapy will entail a solution to such complex disturbances, additionally because some forms of
comorbidity may be covered by the acute manifestations of the disorder and become evident only
when the most severe symptoms have abated.26
Use of psychotherapy
after pharmacological treatment
Instead of administering different therapies together, their sequential administration is planned, based
on some specific effects induced by each of them
that provide additional benefits in the course of
time. Staging has the potential to improve the logic and timing of interventions.11 The phenomenological development of unipolar depression may be
categorized according to stages (Table I).14 A drug
that may be effective in stage 2 major depressive
episode (previously untreated) may not be as effective in a patient with stage 4 recurrent major depression who has been submitted to previous trials.
Most of the patients in STAR*D were in stages 4 or
5 of Table I. Pharmacological manipulations, either
by switching or augmentation (steps 1 and 2), may
propel depressive illness into a refractory phase,
characterized by low remission, high relapse, and
high intolerance (steps 3 and 4).11
In a controlled therapeutic trial,27 40 patients with
major depressive disorder who had been successfully treated with antidepressant drugs were randomly assigned to either CBT or clinical management of
residual symptoms. In both groups, antidepressant
drugs were tapered and discontinued. The group
that received CBT treatment had a significantly lower level of residual symptoms after drug discontinuation in comparison with the clinical management
group. CBT also resulted in a lower rate of relapse,
with achievement of statistical significance at a 4year follow-up.28 These differences faded at a 6-year
follow-up.29 However, when multiple relapses were
considered, patients in the CBT group had a significantly lower number of depressive episodes than
those in the standard clinical management group.29
The aim of this approach was to spend CBT resources when they are most likely to make a unique
Rationale for sequential treatment
and separate contribution to patient wellbeing and
to achieve a more pervasive recovery. This sequenStandard treatment of depression, even in specialtial approach was also applied to 40 patients with
ized settings, seems to yield modest and temporary
recurrent major depression (according to the cribenefits and to leave a large amount of residual
teria outlined by Frank et al30) by the same group of
investigators.31 Patients were randomly assigned to
symptomatology, which appears to be one of the
either CBT for residual symptoms—supplementstrongest predictors of an unfavorable outcome.15,16
These findings have led to the hypothesis that resided by lifestyle modification and wellbeing theraual symptoms upon recovery may progress to bepy32,33 — or clinical management. In both groups,
come prodromal symptoms of relapse, and that
antidepressant drugs were tapered and discontintreatment directed toward residual symptoms may
ued. At a 2-year follow-up, CBT resulted in a signifyield long-term benefits.17 Treatment that is potenicantly lower relapse rate (25%) than did clinical
tially aimed at producing different effects
management (80%). The differential re(eg, pharmacotherapy and psychotheralapse rate was found to be significantly reStages Description
py) may thus be used in a sequential orlated to the abatement of residual sympder. One type of treatment (eg, psychotoms.34 At a 6-year follow-up, CBT still
1
Prodromal phase (anxiety, irritable
resulted in a significantly lower relapse
therapy) may be employed to improve
mood, anhedonia, sleep disorders)
rate (40%) compared with clinical mansymptoms that the other type of treata. No depressive symptoms
agement (90%).35
ment (eg, pharmacotherapy) was unable
b. Minor depression
Other groups of investigators lent supto affect. This may be particularly impor2
Major depressive episode
port to the sequential use of pharmacotant when treatments provide different
therapy and psychotherapy for relapse
modulations of cortical-limbic pathways,
3
Residual phase
prevention in unipolar depression. Paykel
such as CBT and antidepressant drugs in
a. No depressive symptoms
et al36 randomized 158 patients with remajor depression.18
b. Dysthymia
Even though psychotherapy-pharmacent major depression, partially remitted
4
a. Recurrent depression
cotherapy combinations have been shown
with antidepressant treatment but with
b. Double depression
to be more effective than monotherapy
residual symptoms, to clinical managein a number of psychiatric disorders, the
ment or clinical management associated
5
Chronic major depressive episode
effect size observed favoring combined
with cognitive therapy. Patients received
(lasting at least 2 years without
interruptions)
treatment has been generally rather modcontinuation and maintenance antideest in mood and anxiety disorders.19-22 Anpressants during a 1-year follow-up periother line of evidence potentially support- Table I. Stages of primary unipolar depression.14
od. The relapse rate was 47% in the clin-
Increasing remission levels with sequential pharmacotherapy and psychotherapy – Fava and Visani
MEDICOGRAPHIA, VOL 31, No. 2, 2009 187
FOCUS
ical management group and 29% with CBT. There
was a small but statistically significant effect on
residual symptom levels.37 Cost effectiveness analyses showed substantial benefits with the CBT approach.38 At a 6-year follow-up,39 the effects on prevention of relapse and recurrence were found to
persist up to 3 and a half years after the end of CBT.
Similar results were obtained with mindfulnessbased cognitive therapy (MBCT). Teasdale et al40 randomized 145 patients in remission or recovery from
major depression to treatment as usual (TAU) or
TAU supplemented by MBCT. For patients with 3 or
more previous episodes of depression, who constituted 77% of the sample, relapse rates were 66%
for the TAU controls and 37% for the patients also
receiving MBCT.40 However, there were no significant differences in outcome for patients with only
two previous episodes of depression. Since MBCT
was administered in groups, this study provided
the first demonstration that the sequential model
may yield beneficial results also in the group format.
The favorable results concerning MBCT were replicated in a subsequent study.41
In another randomized controlled trial,42 187 patients were randomized to TAU including continuation of pharmacotherapy, or to TAU associated
with group cognitive therapy. During a 2-year follow-up period, cognitive therapy resulted in a significant protective effect, which increased with the
previous number of depressive episodes experienced.
Bockting et al43 identified a significant protective
effect of cognitive therapy in patients with at least
two previous episodes who remitted on various types
of treatments (n=172). Preventive cognitive therapy protected against the influence of a consistently found risk factor for relapse/recurrence (the
number of depressive episodes). This result underlines the potential of psychological preventive interventions.
One study, however, has failed to substantiate the
clinical advantages of the sequential model in unipolar depression.44 A total of 132 patients with major
depression who achieved remission with fluoxetine
were randomized to receive CBT and medication or
medication management alone, and were followed
for up to 28 weeks. Relapse rates did not differ between the two groups, even though the addition of
CBT was associated with attributional style gains.45
A major limitation of this study, however, was the
duration of follow-up (in previous studies, maximal
gains tended to occur at a later point).
The results of the randomized controlled trials
therefore lend support to the use of a sequential
treatment model (pharmacotherapy followed by
psychotherapy) for preventing relapse in unipolar
depression. This approach appears to be particularly important in recurrent depression. However, since
incomplete recovery from the first lifetime major
depressive episode was found to predict a chronic
course of illness during a 12-year prospective naturalistic follow-up study,46 this sequential approach
may be indicated whenever substantial residual
symptomatology is present.
The advantages of keeping patients on their medication during psychotherapy, versus tapering and
188 MEDICOGRAPHIA, VOL 31, No. 2, 2009
discontinuation, have not been directly compared
in sequential studies. Some inferential indications
may come from a study by Blackburn and Moore.47
A total of 75 outpatients with recurrent major depression were allocated to 3 groups: short-term and
maintenance (2 years) treatment with antidepressant drugs, CBT in the short-term and maintenance
phases, and antidepressant use in the short-term
phase with CBT for maintenance. CBT displayed a
similar prophylactic effect to maintenance medication. There were no significant differences among
treatments. These results have been confirmed in a
trial by Hollon et al48 involving 104 patients who responded to treatment (either CBT or medication).
Patients who responded to CBT were withdrawn
from treatment and compared with medication responders who had been randomly assigned to either
continuation medication or placebo withdrawal during a 12-month period. Patients who survived the
continuation phase without relapse were withdrawn
from all treatment and monitored during a 12month naturalistic follow-up period. Patients who
completed CBT were significantly less likely to relapse (31%) than patients on placebo (76%) and no
more likely to relapse than those who carried on
taking continuation medication (47%). Survival
analysis of the naturalistic follow-up indicated that
CBT, unlike antidepressant drugs, had an enduring
effect extending beyond the end of treatment.48 The
results of these two studies47,48 therefore suggest
that discontinuation of antidepressant drugs may
be feasible in subgroups of patients when CBT is
provided.
A novel indication for the sequential model was
provided by a very small pilot study, which concerned 10 patients with recurrent depression who
relapsed while taking maintenance antidepressant
drugs.49 They were randomly assigned to a dose increase and clinical management, or to CBT and
maintenance of the antidepressant drug at the same
dose. Four of five patients responded to a larger
dose, but all had relapsed again at that dose by a
1-year follow-up. Four of five patients responded
to CBT, but only one relapsed during follow-up. In
another recent investigation,50 the feasibility of a
family intervention approach to the loss of clinical
effect during long-term antidepressant therapy was
explored. A total of 20 outpatients with recurrent
major depressive disorder who lived with a partner
and had relapsed while taking antidepressant drugs
were randomly assigned to (i) family intervention
approach according to the McMaster Model and
maintenance of the antidepressant drug at the same
dosage or (ii) dose increase and clinical management. Seven out of 10 patients responded to an increased dosage; all but 1 relapsed again on that
dosage during follow-up. Seven out of 10 patients
responded to family intervention, but only 1 relapsed during follow-up. The data from these pilot
studies49,50 have to be confirmed with large-scale
controlled studies, but may suggest that the application of a sequential model is feasible when there
is a loss of clinical effect during long-term antidepressant treatment.2 Treatment for patients with
psychotic depression is urgently needed due to the
Increasing remission levels with sequential pharmacotherapy and psychotherapy – Fava and Visani
FOCUS
increased risk of morbidity and mortality associated with this population.51 Patients with psychotic
depression show a poorer response to antidepressant treatment with tricyclics or selective serotonin
reuptake inhibitors alone compared with nonpsychotic depressed individuals.52,53
Pilot data by Gaudiano et al51 recently suggested
that patients with psychotic depression can benefit from modified CBT approaches (acceptance and
commitment therapy, ACT). In this pilot trial, 40 patients were randomly assigned to enhanced treatment as usual (ETAU) or ETAU plus individual ACT
sessions. ACT was associated with clinically significant reductions in acute symptom severity and impairment compared with treatment as usual. Gaudiano et al51 found it appropriate to begin therapy
with most hospitalized patients once they were able
to participate in other group therapy on the unit,
after initial psychiatric stabilization with medications. After acute psychotic symptoms dissipate and
antipsychotic medication is withdrawn, psychotherapy can be started to help patients monitor residual
symptoms and learn to cope more effectively with
stressors to prevent symptom recurrence.
Use of pharmacotherapy
after psychological treatment
There is little research on the sequential use of psychotherapy and pharmacotherapy, despite the fact
that successful psychotherapy is also associated
with substantial residual symptomatology.15 Frank
et al54 used a successive cohort approach to compare
two similar groups of female patients with recurrent
unipolar depression: one in which the combination
of interpersonal psychotherapy (IPT) and pharmacotherapy was initiated at the beginning (n=180),
and a second in which IPT alone was first provided, with only those not remitting being given the
combination treatment (n=159). The remission rate
was significantly higher in the latter group. The
results thus suggest that the strategy of offering
IPT to women with recurrent depression, adding
pharmacotherapy only in the case of incomplete remission, might be advantageous.
Sequential use of
two pharmacological strategies
The sequential use of pharmacological strategies
in affective disorders has been traditionally limited to instances of treatment resistance.2 A notable
exception has been the use of lithium to reduce relapse in unipolar depression.55
It has also been suggested56 that the most effective drugs in treating acute depression may not be
the most suitable for post-acute or continuation
treatment. During a 6-year follow-up in a randomized trial comparing the sequential use of pharmacotherapy and cognitive behavioral treatment
versus clinical management in patients with recurrent depression,35 no antidepressant drugs were used
unless a relapse ensued. Patients were then treated
with the same antidepressant drug that had been
used in the previous episode. Clonazepam was added
Increasing remission levels with sequential pharmacotherapy and psychotherapy – Fava and Visani
to the treatment regimen and continued when the
antidepressant drug was stopped. The mean survival
time after introduction of clonazepam was significantly longer than that before the first relapse.
Sequential use of
two psychotherapeutic techniques
Current psychotherapeutic strategies, particularly
in the CBT realm, use several elements from the
beginning (eg, cognitive restructuring, exposure,
relaxation, etc). It would be of interest to verify
whether the sequential use of single treatment
components can yield significant advantages, both
in mood and anxiety disorders. For instance, in a
modified CBT approach to drug-resistant major depression,4 therapeutic ingredients were introduced
at different times (behavioral activation first and
cognitive restructuring later).
Implications for assessment
and treatment planning
The literature that has been reviewed here has potential implications for clinical practice, but it
should be interpreted with caution in view of several issues. First of all, there are insufficient studies
exploring the various types of sequential approach
in unipolar depression, with the exception of pharmacotherapy followed by psychotherapy. Further,
the sequential design is exposed to the risk that one
treatment is provided in a more expert manner than
another (eg, the use of psychotropic drugs compared with psychotherapy). Third, certain types of
treatment that have been used, such as mindfulness
therapy and wellbeing therapy, may not be widely
available, and there are difficulties entailed in their
translation from the “expert” site to clinical practice.
Fourth, when the results of a sequential treatment
are compared with those of a minimal intervention
control group (such as clinical management or
TAU), there is the possibility that the treatment effect may have been achievable with any active treatment and may not be specific to the treatment at
hand. Finally, several of the aforementioned studies were based around patients who had responded
to initial treatment, and this might have led to an
undervaluation of the fact that patients at high risk
may have dropped out early.
Nonetheless, there are considerable implications
for assessment and treatment planning that are
worthy of clinical attention. The sequential model
calls in fact for a substantial modification of the
flat, cross-sectional approach based on DSM-IV (Diagnostic and Statistical Manual of Mental Disorders–Fourth Edition) criteria only, which ignores
the longitudinal development of depressive illness,
previous episodes, and responses to previous treatments.3 A satisfactory assessment requires multiple points of observation during the course of affective illnesses. Such observations may disclose
psychopathological features that are overshadowed
by the acute manifestations of the affective disorder.
As a result, three assessment phases are required,
with modalities that depart from those commonly
MEDICOGRAPHIA, VOL 31, No. 2, 2009 189
FOCUS
used in psychiatric practice. The key issue is in fact
to match treatment ingredients with psychopathological findings.
Initial assessment
The majority of patients with mood and anxiety disorders do not qualify for one, but for several axis I
and axis II disorders.23,24 However, there is comorbidity that wanes upon successful treatment of depression and comorbidity that persists, in syndromic
or subsyndromic forms (residual symptoms). Clinical differentiation of such morbidity requires a shift
from the current psychometric model (where severity is determined by the number of symptoms and
not by intensity or quality) to a clinimetric model,57 which may allow a definition of the progression, extent, and severity of depressive illness.
As a response to the current flat diagnostic evaluation, Emmelkamp et al58 have introduced the
concept of macro-analysis (a relationship between
co-occurring syndromes is established on the basis of where treatment should commence first). The
planning of sequential treatment thus requires determination of the symptomatic target of the firstline approach (eg, pharmacotherapy), and tentative
identification of other areas of concern to be addressed by subsequent treatment (eg, psychotherapy). Organization of different DSM syndromes by
macro-analysis is thus the key to successful implementation of the sequential model.1
Re-assessment after the first line of treatment
has been completed
It is of the utmost importance that the patient be
reassessed after the first line of treatment has been
completed, in order to establish the level of remission in the patient and whether residual symptoms
are occurring and further treatment is necessary.59
In two studies27,31 concerned with the sequential
treatment of depression, reassessment was performed after 3 months of drug treatment, when
maximal benefits were likely to be present.60 There
are several major obstacles to a satisfactory assessment of the patient at this stage. The first lies in the
exploration of only a few target symptoms, instead
of the full spectrum of psychopathology (as if he or
she were a new patient). The second pitfall derives
from the fact that the hidden conceptual model in
clinical assessment is psychometric, in which severREFERENCES
1. Fava GA, Ruini C, Rafanelli C. Sequential treatment
of mood and anxiety disorders. J Clin Psychiatry.
2005;66:1392-1400.
2. Fava GA. Can long-term treatment with antidepressant drugs worsen the course of depression? J Clin
Psychiatry. 2003;64:123-133.
3. Stewart JW, Mercier MA, Agosti V, Guardino M,
Quitkin FM. Imipramine is effective after unsuccessful cognitive therapy. J Clin Psychopharmacology.
1993;13:114-119.
4. Fava GA, Savron G, Grandi S, Rafanelli C. Cognitive
behavioral management of drug-resistant major depressive disorder. J Clin Psychiatry.1997;58:278-282.
5. Rush AJ, Trivedi MH, Wisniewski SR, et al. Acute
and longer-term outcomes in depressed outpatients
requiring one or several treatment steps. Am J Psychiatry. 2006;163:1905-1917.
6. Fava M, Evins AE, Dorer DJ, Schoenfeld DA. The
190 MEDICOGRAPHIA, VOL 31, No. 2, 2009
ity is determined by the number of symptoms, not
by their intensity or quality. Further, the assessment of subclinical symptomatology, as frequently
occurs in the setting of remitted or partially remitted disorders,16,18 cannot exclude consideration of
the symptoms’ longitudinal development (prodromal phase, the fully developed disorder, and residual states). Detre and Jarecki 61 provided a model for
relating prodromal and residual symptomatology
in psychiatric illness, defined as the rollback phenomenon: as the illness remits, it progressively recapitulates (though in a reverse order) many of the
stages and symptoms that were seen during the
time it developed. Finally, in clinical practice as well
as in research, collection of symptom information is
performed during a clinical interview. However, selfobservation, in which the patient is instructed to
report in a diary the most important episodes of distress that may have ensued in a specific time period,
such as a couple of weeks, is an important source
of information concerned with allostatic load.33
Final assessment after the second line of
treatment has been completed
Final assessment should take place after the second line of treatment has been completed; for instance, in a depressed patient when psychotherapy
following pharmacotherapy has been performed
and medications have been discontinued.59 If substantial residual symptomatology persists despite
clinical response, then new treatment strategies,
such as long-term, indefinite drug therapy, should
be discussed with the patient.
Conclusion
The sequential treatment of mood and anxiety disorders does not fall within the realm of maintenance
strategies, which have the aim of prolonging clinical responses obtained with treatments.62 It is an intensive, two-stage approach, which derives from the
awareness that one course of treatment with a specific tool (whether pharmacotherapy or psychotherapy) is unlikely to entail a solution to the affective
disturbances of patients. The aim of the sequential
approach is to add therapeutic ingredients as long
as they are needed. Therapeutic targets are not predetermined, but depend on the response of patients
to the first course of treatment. problem of the placebo response in clinical trials for
psychiatric disorders. Psychother Psychosom. 2003;
72:115-127.
7. Vieta E, Carnè X. The use of placebo in clinical trials on bipolar disorder. Psychother Psychosom. 2005;
74:10-16.
8. Gaudiano BA, Herbert JD. Methodological issues
in clinical trials of antidepressant medications. Psychother Psychosom. 2005;74:17-25.
9. Thase ME, Friedman ES, Biggs MM, et al. Cognitive therapy versus medication in augmentation and
switch strategies as second-step treatments: a STAR*D
report. Am J Psychiatry. 2007;164:739-752.
10. Nelson JC. The STAR*D study: a four course meal
that leaves us wanting more. Am J Psychiatry. 2006;
163:1864-1866.
11. Fava GA, Tomba E, Grandi S. The road to recovery from depression. Don’t drive today with yesterday’s
map. Psychother Psychosom. 2007;76:260-265.
12. Fava GA, Kellner R. Staging: a neglected dimension in psychiatric classification. Acta Psychiatr Scand.
1993;87:225-230.
13. McGorry PD, Hichie IB, Yang AR, Pantelis C,
Jackson HJ. Clinical staging of psychiatric disorders.
Aust N Zeal J Psychiatry. 2006;40:616-622.
14. Fava GA, Tossani E. Prodromal stage of major depression. Early Interv Psychiatry. 2007;1:9-18.
15. Fava GA. Subclinical symptoms in mood disorders. Psychol Med. 1999;29:47-61.
16. Fava GA, Ruini C, Belaise C. The concept of recovery in major depression. Psychol Med. 2007;37:
307-317.
17. Fava GA, Kellner R. Prodromal symptoms in affective disorder. Am J Psychiatry. 1991;148:823-830.
18. Goldapple K, Segal Z, Garson C, et al. Modulation
of cortical-limbic pathways in major depression. Arch
Increasing remission levels with sequential pharmacotherapy and psychotherapy – Fava and Visani
FOCUS
Gen Psychiatry. 2004;61:34-41.
19. Thase ME, Jindal RD. Combining psychotherapy
and psychopharmacology for treatment of mental
disorders. In: Lambert MJ, ed. Bergin and Garfield’s
Handbook of Psychotherapy and Behavior Change.
New York, NY: Wiley; 2004:743-766.
20. Otto MW, Smits JAJ, Reese HE. Combined psychotherapy and pharmacotherapy for mood and anxiety
disorders in adults. Clin Psychol Sci Pract. 2005;12:
72-86.
21. Hollon SD, Jarrett RB, Nierenberg AA, Thase ME,
Trivedi M, Rush AJ. Psychotherapy and medication in
the treatment of adult and geriatric depression: which
monotherapy or combined treatment? J Clin Psychiatry. 2005;66:455-468.
22. Blom MBJ, Jonker K, Dusseldorp E, et al. Combination treatment for acute depression is superior
only when psychotherapy is added to medication. Psychother Psychosom. 2007;76:289-297.
23. Maj M. The aftermath of the concept of psychiatric
comorbidity. Psychother Psychosom. 2005;74:65-67.
24. Pincus HA, Tew JD, First MB. Psychiatric comorbidity: is more less? World Psychiatry. 2004;3:18-23.
25. Zimmerman M, Chelminski I, McDermut W. Major depressive disorder and Axis I diagnostic comorbidity. J Clin Psychiatry. 2002;63:187-191.
26. Gaynes BN, Magruder KM, Burns BJ, Wagner HR,
Yarnall KSH, Broadhead WE. Does a coexisting anxiety disorder predict persistence of depressive illness
in primary care patients with major depression? Gen
Hosp Psychiatry. 1999;21:158-167.
27. Fava GA, Grandi S, Zielezny M, Canestrari R, Morphy MA. Cognitive behavioral treatment of residual
symptoms in primary major depressive disorders. Am
J Psychiatry. 1994;151:1295-1299.
28. Fava GA, Grandi S, Zielezny M, Rafanelli C, Canestrari R. Four year outcome for cognitive behavioral
treatment of residual symptoms in major depression.
Am J Psychiatry. 1996;153:945-947.
29. Fava GA, Rafanelli C, Grandi S, Canestrari R, Morphy MA. Six year outcome for cognitive behavioral
treatment of residual symptoms in major depression.
Am J Psychiatry. 1998;155:1443-1445.
30. Frank E, Kupfer DJ, Perel JM, et al. Three year
outcomes for maintenance therapies in recurrent depression. Arch Gen Psychiatry. 1990;47:1093-1099.
31. Fava GA, Rafanelli C, Grandi S, Conti S, Belluardo
P. Prevention of recurrent depression with cognitive
behavioral therapy. Arch Gen Psychiatry. 1998;55:
816-820.
32. Fava GA, Ruini C. Development and characteristics of a well-being enhancing psychotherapeutic
strategy: well-being therapy. J Behav Ther Exp Psychiatry. 2003;34:45-63.
33. Fava GA, Ruini C. The sequential approach to re-
lapse prevention in unipolar depression. World Psychiatry. 2002;1:10-15.
34. Fava GA, Rafanelli C, Grandi S, Conti S, Belluardo
P. The role of residual subthreshold symptoms in early episode relapse in unipolar major depressive disorder. Arch Gen Psychiatry. 1999;56:765.
35. Fava GA, Ruini C, Rafanelli C, Finos L, Conti S,
Grandi S. Six-year outcome of cognitive behavior therapy for prevention of recurrent depression. Am J Psychiatry. 2004;161:1872-1876.
36. Paykel ES, Scott J, Teasdale JD, et al. Prevention
of relapse in residual depression by cognitive therapy.
Arch Gen Psychiatry. 1999;56:829-835.
37. Scott J, Teasdale JD, Paykel ES, et al. Effects of
cognitive therapy on psychological symptoms and social functioning in residual depression. Br J Psychiatry. 2000;170:440-446.
38. Scott J, Palmer S, Paykel ES, Teasdale JD, Hayhurst H. Use of cognitive therapy for relapse prevention in chronic depression: cost effectiveness study.
Br J Psychiatry. 2003;182:221-227.
39. Paykel ES, Scott J, Cornwall PL, et al. Duration of
relapse prevention after cognitive therapy in residual depression: follow-up of controlled trial. Psychol
Med. 2005;35:59-68.
40. Teasdale JD, Segal ZV, Williams JMG, Ridgeway
VA, Soulsby JM, Lan MA. Prevention of relapse/recurrence in major depression by mindfulness-based cognitive therapy. J Consult Clin Psychol. 2000;68:615623.
41. Ma SH, Teasdale JD. Mindfulness-based cognitive
therapy for depression. J Consult Clin Psychol. 2004;
72:31-40.
42. Bockting CLH, Schene AH, Spinhoven P, et al.
Preventing relapse/recurrence in recurrent depression
using cognitive therapy. J Consult Clin Psychol. 2005;
73:647-657.
43. Bockting CLH, Spinhoven P, Koeter MVT, Wounters LF, Schene AH. Differential predictors of response
to preventive cognitive therapy in recurrent depression. Psychother Psychosom. 2006;75:229-236.
44. Perlis RH, Nierenberg AA, Alpert JE, et al. Effects
of adding cognitive therapy to fluoxetine dose increase
on risk of relapse and residual depressive symptoms
in continuation treatment of major depressive disorder. J Clin Psychopharmacol. 2002;22:474-480.
45. Petersen T, Harly R, Papakostas G, Montoya HD,
Fava M, Alpert JE. Continuation cognitive behavioral
therapy maintains attributional style improvement in
depressed patients responding acutely to fluoxetine.
Psychol Med. 2004;34:555-561.
46. Judd LJ, Paulus MJ, Schettler PJ, et al. Does incomplete recovery from first lifetime major depressive
episode herald a chronic course of illness? Am J
Psychiatry. 2000;157:1501-1504.
47. Blackburn IM, Moore RG. Controlled acute and
follow-up trial of cognitive therapy in outpatients with
recurrent depression. Br J Psychiatry. 1997;171:328334.
48. Hollon SD, de Rubeis RJ, Shelton RC, et al. Prevention of relapse following cognitive therapy vs medications in moderate to severe depression. Arch Gen
Psychiatry. 2005;62:417-422.
49. Fava GA, Ruini C, Rafanelli C, Grandi S. Cognitive
behavior approach to loss of clinical effect during
long-term antidepressant treatment. Am J Psychiatry.
2002;159:2094-2095.
50. Fabbri S, Fava GA, Rafanelli C, Tomba E. Family
intervention approach to loss of clinical effect during
long-term antidepressant treatment: a pilot study.
J Clin Psychiatry. 2007;68:1348-1351.
51. Gaudiano BA, Miller IW, Herbert JD. The treatment of psychotic major depression: is there a role for
adjunctive psychotherapy? Psychother Psychosom.
2007;76:271-277.
52. Vega JAW, Mortimer AM, Tyson PJ. Somatic treatment of psychotic depression: review and recommendations for practice. J Clin Psychopharmacol. 2000;
20:504-519.
53. Tyrka AR, Price LH, Mello MF, Mello AF, Carpenter LL. Psychotic major depression: a benefit-risk assessment of treatment options. Drug Saf. 2006;29:
491-508.
54. Frank E, Grochocinski VJ, Spanier CA, et al. Interpersonal psychotherapy and antidepressant medication: evaluation of a sequential treatment strategy
in women with recurrent major depression. J Clin
Psychiatry. 2000;61:51-57.
55. Davies JM, Janicak PG, Hogan DM. Mood stabilizers in the prevention of recurrent affective disorders. Acta Psychiatr Scand. 1999;100:406-417.
56. Fava GA. The concept of recovery in affective disorders. Psychother Psychosom. 1996;65:2-13.
57. Fava GA, Ruini C, Rafanelli C. Psychometric theory is an obstacle to the progress of clinical research.
Psychother Psychosom. 2004;73:145-148.
58. Emmelkamp PMG. The additional value of clinimetrics needs to be established rather than assumed.
Psychother Psychosom. 2004;73:142-144.
59. Fava GA, Ruini C, Sonino N. Management of recurrent depression in primary care. Psychother Psychosom. 2003;72:3-9.
60. Keller MB, Lavori PW, Mueller TI, et al. Time to
recovery, chronicity and levels of psychopathology
in major depression. Arch Gen Psychiatry. 1992;49:
809-816.
61. Detre TP, Jarecki H, eds. Modern Psychiatric
Treatment. Philadelphia, Pa: Lippincott; 1971.
62. Kupfer DJ. Maintenance treatment in recurrent
depression. Br J Psychiatry. 1992;161:309-316.
AUGMENTATION DU NIVEAU DE RÉMISSION DANS LA DÉPRESSION
PAR UTILISATION SÉQUENTIELLE DE LA PHARMACOTHÉRAPIE ET DE LA PSYCHOTHÉRAPIE
L
e sombre avenir à long terme de la dépression en termes
de rechute et de récidive est de plus en plus documenté.
Cette évolution insatisfaisante semble liée à l’existence
d’une symptomatologie résiduelle importante. Si les symptômes
résiduels sont la règle à la conclusion d’un traitement pharmacologique ou psychothérapeutique et que leur présence est corrélée à un mauvais pronostic (du fait qu’ils peuvent évoluer après
la guérison en symptômes prodromiques d’une rechute), les traiter peut s’avérer bénéfique à long terme. Les traitements visant
des symptômes potentiellement différents, comme la pharmacothérapie et la psychothérapie, pourraient être utilisés dans un
ordre séquentiel, l’un (par exemple la psychothérapie) employé
pour améliorer les symptômes que l’autre (par exemple la pharmacothérapie) ne peut soigner. Plusieurs études étayent les avan-
tages cliniques de l’utilisation séquentielle de la psychothérapie après la pharmacothérapie. Peu de recherches ont été faites
sur les autres formes de traitement séquentiel de la dépression.
Les médicaments les plus efficaces dans le traitement d’attaque
de la dépression ne sont pas nécessairement les plus appropriés
pour le traitement de la phase de consolidation ou d’entretien.
Le traitement séquentiel de la maladie dépressive n’appartient
pas aux stratégies de maintenance. Il s’agit d’une approche intensive en deux étapes basée sur le fait qu’un type de traitement
spécifique (pharmacothérapie ou psychothérapie) ne peut détenir la solution de l’ensemble complexe des symptômes présentés par les patients dépressifs. Le modèle séquentiel a des implications considérables pour l’évaluation et la planification du
traitement.
Increasing remission levels with sequential pharmacotherapy and psychotherapy – Fava and Visani
MEDICOGRAPHIA, VOL 31, No. 2, 2009 191
A
T
O U C H
O F
F
R A N C E
GU Qian (顾倩), MD
NI Lidong (倪黎冬), MD
HU Weiguo (胡伟国), MD
Hospital Office, Ruijin
Hospital, Shanghai, CHINA
Ruijin Hospital
From 19th-century French Jesuit Hospital
to present-day state-of-the-art
Chinese University Hospital
瑞金医院
从19世纪法国耶稣会医院到当今中国先进的现代化医院
by GU Qian, NI Lidong, and
HU Weiguo, China
R
Banners for the
Centennial Anniversary
of Guangci/Ruijin
Hospital in Shanghai
(1907-2007). “Show
great kindness, pursue
excellence” reads the
motto on the right-hand
banner. Courtesy of
Ruijin Hospital.
uijin Hospital (瑞金医院), which originated as the medical wing of Aurora University
(震旦大学, literally “Thunder of Sunrise”), a French-Catholic institution founded in Shanghai in 1903 and dissolved in 1952, is today affiliated to the Shanghai Jiao Tong University
School of Medicine (上海交通大学医学院). Despite changes of name—it began life as l’Hôpital
Sainte-Marie, then became Guangci Hospital (廣慈医院, literally, “Great Mercy Hospital”) —
it retains vigorous reminders of its French past during the first half of the 20th century when
most of its doctors, whether French or Chinese nationals, were not only fluent French speakers but the
respected and expert practitioners of a French-accented medicine. The history of Aurora University, its
medical school, and hospital provides a fascinating glimpse of enlightened European and Catholic outreach in a largely impoverished oriental metropolis during the early 1900s. It is also a striking example,
A
www.medicographia.com
Address for correspondence:
Dr HU Weiguo, MD, Hospital
Office, Ruijin Hospital,
197 Ruijin 2nd Road,
Shanghai 200025, China.
(e-mail: wghusurgeon@
hotmail.com)
urora Academy (Zhendan Xueyuan 震旦学院, literally, “Thunder of Sunrise”) was founded in
Shanghai in 1903 by a Chinese Jesuit, Joseph Ma Xiangbo (马相伯), as a French-Catholic institution whose mission was to modernize his country. In 1907, the Jesuits acquired nearby land on
which they built l’Hôpital Sainte-Marie, later known as Guangci (廣慈医院) Hospital—literally, “Great Mercy Hospital.” It became a teaching hospital for the Academy’s medical students.
In 1917, the Academy achieved university status, supplying doctors to the Catholic hospitals of
China, and also training nurses and medical technicians. Under a succession of French Jesuit principals
it was a beacon of French language and French medicine, practiced by a mixture of French and Chinese
nationals. In 1952, with the closure of independent educational institutions throughout China, Aurora
University merged into Fudan University (复旦大学), while its medical school merged into Shanghai Second Medical College (上海第二医学院). Guangci became Ruijin Hospital (瑞金医院) and developed into
one of China’s leading university hospitals. In the 1980s, Shanghai Second Medical College reconnected
with its past by offering a 7-year course in French language and medicine. Ever since there has been a vigorous exchange program between the two countries at every level, personified by Ruijin’s internationally renowned hematologist Chen Zhu (陈竺), who followed his postgraduate studies at Shanghai Second
Medical College with a doctorate in Paris and is now China’s minister of health. With increasing numbers
of French physicians and scientists coming to Ruijin Hospital to learn rather than to teach, the hopes that
a French-trained Chinese Jesuit pinned on his fledgling Aurora Academy have finally been realized.
Medicographia. 2009;31:198-204.
198 MEDICOGRAPHIA, VOL 31, No. 2, 2009
(see French abstract on page 204)
Ruijin Hospital – GU Qian, NI Lidong, and HU Weiguo
A TOUCH
OF
FRANCE
Ruijin Hospital
continues Guangci Hospital
(Hôpital Sainte-Marie).
Bottom left of the picture,
one can see the low
red-tiled brick buildings of
the former French hospital.
Courtesy of Ruijin Hospital.
within the medical sphere, of accommodation and cross-fertilization between two ancient and contrasting cultures, those of France and China. The founder of Aurora University was a Chinese Jesuit, Joseph
Ma Xiangbo (马相伯) (1860-1939). Born into a well-to-do Catholic family, his early education was in the
Chinese classics. At the age of 12, he was enrolled in St Ignatius College, newly opened by the French Jesuits in Shanghai. After ordination at the age of 30, he returned to St Ignatius as its principal. Excelling
in astronomy, mathematics, and Latin, he set out to translate European scientific works into Chinese. In
1876, however, he left the priesthood, married, and spent the next 21 years working in business and government on behalf of the Chinese Self-Strengthening Movement that advocated learning from the West.
In that capacity he traveled abroad, not only to Korea but also to the United States and Europe. But as the
Movement waned and after his wife died, Ma Xiangbo rejoined the Jesuits in Shanghai in 1898 and five
years later opened the Aurora Academy (Zhendan Xueyuan [震旦学院]) with the purpose of furthering his
original aim of translating the Western books that China needed in its drive toward modernization, and he
chose its name, “Thunder of Sunrise,” to mark the Chinese new dawn that he hoped it would usher in.
In 1908, Aurora University introduced a French-style undergraduate curriculum
including a three-year preparatory program and a three-year advanced program. The
preparatory program included subjects such as French, English, history, philosophy,
geography, elementary mathematics, physics, and other natural sciences. On successful completion of this program, students opted between the four majors of the advanced
program: humanities, law, medical science, and philosophy. From grade 2 of the preparatory program upwards, all courses except Chinese were taught in French.
Meanwhile the Jesuit Apostolic Vicar of nearby Nanking, Próspero París (姚宗李)
(1846-1931), had bought 165 acres of Shanghai land east of the main thoroughfare
Jinshenpu Lu 金神圃路 (modern Ruijin No. 2 Avenue [瑞金二路]) in order to found
a hospital. The first construction phase consisted of four Western-style two-floor brickand-timber houses, two of which contained wards for a total of 100 patients. The opening ceremony took place on October 13, 1907. Known initially to Europeans as the “Hôpital Sainte-Marie,” it was renamed Guangci Hospital (廣慈医院) several years later.
French-trained Jesuit priest Ma
Xiangbo (马相伯, 1840-1939),
Aurora Academy selected a site near the hospital to found a medical school. Its stufounder of Aurora Academy
dents could do their ward work in the hospital, and the most outstanding graduates
in Shanghai in 1903, which
were selected as interns. In those early days, two French doctors managed the inpatients
lives on today its merging with
Shanghai Jiaotong University.
while nuns cared for the outpatients, from examination through diagnosis to treatment
© Culture Sino-France, with
and dispensing. Wards were arranged hierarchically (missionary ward, first-class male
kind permission.
and female wards, second-class civilian wards) and by specialty (obstetrics, isolation, etc).
There were also special wards, such as for French armed forces personnel, police officers from Annam (the
French protectorate in central Vietnam), and criminals.
The first operating rooms opened in 1918 and catered for three or four major cases daily, primarily in
gastrointestinal surgery (hemorrhoids, appendix, bowel, stomach, and biliary tract), but also urinary tract
and gynecological surgery. In 1921, Guangci Hospital became one of the first in China to open a department of radiology. Its Pasteur Building housed some celebrated laboratories. The French Concession’s
Ruijin Hospital – GU Qian, NI Lidong, and HU Weiguo
MEDICOGRAPHIA, VOL 31, No. 2, 2009 199
A TO
UCH
OF
FRA
NCE
Still extant maternity
(“Maternité”) ward of
former Hôpital SainteMarie, founded by French
Jesuits in 1907, now part
of the modern Ruijin
Hospital. Courtesy of
Ruijin Hospital.
Zhendan Daxue
(Aurora University)
building in 1939.
© Culture Sino-France,
with kind permission.
Industry Bureau funded the five-floor St Vincent de Paul building containing 300 beds. The board of directors, set up in the 1930s, included the French consul general, the heads of the Industry Bureau, and
relevant Church dignitaries. In 1936, the Hospital founded a nursing school for specialist nurses and medical technicians. By 1940, when Guangci Hospital registered with the Shanghai government, it had become
a 780-bed institution and the largest hospital in the Far East.
Having started out as wholly French-Catholic in inspiration and practice, deploying exclusively French
treatments dispensed by French doctors and nuns, Guangci Hospital evolved over the years to absorb more
and more influence from its Chinese environment. It became a Sino-French institution hosting an uninterrupted and indeed increasing interchange
of science and talent that attracted — and continues to attract—some of China’s best medical
brains onto both its faculty and student body.
In 1933, Dr Kuang Ankun (邝安堃), who had
obtained his medical degree in Paris, returned
home as professor of internal medicine at Aurora University and chief of internal medicine
at Guangci Hospital. He transformed the theory
and practice of his specialty at the University. His
immediate concerns were the prevention and
treatment of diseases such as relapsing fever, typhoid, and amebic dysentery, and he was among
the first in China to diagnose lupus erythematosus and temporal arteritis, attracting considerable public interest at the time. He was followed
in 1935 by Dr Xu Baoyi (徐宝彝), a surgeon with
degrees from the University of Lyon, who returned home to become professor of surgical science at Aurora
University and chief of surgery at Guangci Hospital. His skill in thyroid and gastrointestinal surgery, including surgery of the biliary tract, earned him a regional and international reputation. He was elected
to the International Society of Surgery and chaired the Shanghai Medical Association.
An impressive number of French physicians and surgeons subsequently joined the Aurora University
faculty and served on the staff of Guangci Hospital. Their specialties covered surgery, pediatrics, infectious
disease, and radiology. Working alongside French or Belgian-trained Chinese nationals, such as the surgeon Dr Fu Peibin (傅培彬), the obstetrician and gynecologist Dr Tang Shiheng (唐士恒), the urologist
Dr Cheng Yixiong (程一雄), the otorhinolaryngologist Dr Liu Tao (刘涛), and the dermatologist Dr Zhu
Zhonggang (朱仲刚), they laid the foundation for the Hospital’s future development.
In 1952, all the faculties of Aurora University (震旦大学), including its medical school, were combined
with the faculties of Fudan (復旦), which Ma Xiangbo had helped establish, East China Normal University
(华东师范大学), and Shanghai Second Medical College (上海第二医学院). Guangci Hospital (廣慈医院)
became the College’s affiliated teaching hospital. The College was renamed Shanghai Second
Medical University (上海第二医科大学) in 1985,
and in 2005 was merged with Shanghai Jiao
Tong University (上海交通大学) to be renamed
Shanghai Jiao Tong University School of Medicine (上海交通大学医学院). Guangci Hospital itself was renamed Dong Fang Hong Yiyuan
(“The East is Red” Hospital, 东方红医院) in 1966,
then Ruijin Hospital (瑞金医院) in 1972.
In the 1980s, Shanghai Second Medical University reconnected with its roots by recruiting
students for a 7-year course in French language
and medicine, the first of its kind in China. It has
since contracted with some 18 French universities and medical schools to host exchange students and teaching faculty in multiple medical disciplines. Up to two thirds of the University’s foreign
exchange medical students have studied in France. In 1996, Ruijin Hospital agreed a staff training and
academic exchange scheme with the Paris university hospital authorities. One of the aims spelled out in
the Sino-French health cooperation agreement announced by French President Jacques Chirac during
his visit to China, when he came to Ruijin Hospital, was to support the French medicine program. Students
200 MEDICOGRAPHIA, VOL 31, No. 2, 2009
Ruijin Hospital – GU Qian, NI Lidong, and HU Weiguo
A TOUCH
Prof Cheng Yixiong
(程一雄) (1912-2006).
Urologist. Graduated
from Aurora University
with a PhD in Medicine,
specialized in urological
surgery in Belgium and
France. Participated in
the establishment of the
Shanghai Second Medical
College and set up the
Department of Urological
Surgery of Ruijin Hospital,
where he was a tenured
professor. Courtesy of
Ruijin Hospital.
Prof Chen Zhu (陈竺).
Masters Degree at
Shanghai Second Medical
College, 1989, PhD at
Paris No. 7 University.
Member of the Chinese
Academy of Sciences,
tenured professor at
Ruijin Hospital. 1981,
Director of Shanghai
Hematological Institute.
Major contributions to
the knowledge of
leukemia, with findings
published in international
journals. Established the
State South Research
Center on the Human
Genome. He is China’s
current Minister of
Health. Courtesy of Ruijin Hospital.
Prof Chen Saijuan
(陈赛娟). Member of
the Chinese Academy of
Engineering, tenured
professor at Ruijin
Hospital. 1971, MD at
Shanghai Second Medical
University, 1981, PhD
at Paris No. 7 University.
At present Executive
Director of Shanghai
Hematology Institute,
Director of the of Medical
Genome Laboratory.
Courtesy of Ruijin
Hospital.
OF
FRANCE
Prof Wang Zhenyi
(王振义). Tenured professor at Ruijin Hospital.
Graduated with an MD and
PhD (1948) at Aurora
University Medical College.
After working at Guangci
Hospital, participated in
the establishment of the
Shanghai Second Medical
College of which he
became president. At present honorary director of
Shanghai Hematological
Institute. Ham-Wasserman
Lecture Prize in 2003,
Foreign Member of French
Academy of Science, and
Honorary PhD in Science
at Columbia University,
USA. Courtesy of Ruijin
Hospital.
on the program not only complete a broader introductory course in the humanities and social sciences,
they also receive a firmer foundation in the natural sciences, a more solid grounding in basic medical theory and a more systematic introduction to clinical medicine. At the same time, they become proficient in
French, to the extent that they can read French medical documents and communicate in the language.
Half the students on the program are given the opportunity to work for periods as hospital doctors in France
with all costs borne by the host country.
The program faculty combines staff from the Ruijin Hospital School of Clinical Medicine, notably Drs
Wang Zhenyi (王振义), Chen Zhu (陈竺), and other members of the Chinese Academy of Sciences, with
teachers from France, in particular during the first year, when the curriculum comprises no fewer than
726 hours of lectures and exercises in basic French. Between 1997 and 2007, France sent over some 120
MD diploma delivered by Aurora University in 1949.
© 2009, china-news.org (2001)/chine-informations.com
(2005). All rights reserved.
Detail of map of Shanghai (1928) showing Aurora University (red circle) and Sainte Marie (Guangci) Hospital
(green circle). On the upper right, the Chinese Old City
(recognizable by its circular shape) and Huangpu river
(right top and bottom corners). All rights reserved.
Ruijin Hospital – GU Qian, NI Lidong, and HU Weiguo
MEDICOGRAPHIA, VOL 31, No. 2, 2009 201
A TO
UCH
OF
FRA
NCE
Layout of present-day
Ruijin Hospital in
Shanghai. Courtesy
Shanghai Ruijin Hospital.
teaching staff from a wide range of university hospitals. The Ruijin School of Clinical Medicine has also
edited and published the French version of major textbooks in medicine and surgery. In conjunction with
universities in Lyon and Paris, it has set up the SPIRAL resource server to enable its students to access
exactly the same teaching programs and course materials, including interactive video and audio lectures,
to the extent that its students are virtually sitting alongside their French counterparts.
Today Ruijin Hospital is a 1600-bed general teaching hospital occupying a 30-acre site with a floor area
of 60 acres and over 3000 staff, including some nationally and internationally known figures such as Dr
Chen Zhu (陈竺), but also Drs Wang Zhenyi (王振义) and Chen Saijuan (陈赛娟), both members of the
Chinese Academy of Engineering. In addition to its 34 clinical departments, the Hospital has nine departments of medical technology. It also encompasses six Shanghai institutes (Traumatology and Orthopedics,
Hypertension, Endocrine and Metabolic Diseases, Burns, Hematology, and Digestive Tract Surgery), plus
two of the Ministry of Health’s Key Laboratories (Human Genome, and Endocrine and Metabolic Diseases).
Ruijin Hospital has been renowned for its expertise in treating major burns ever since saving steelworker
Qiu Caikang (邱财康) in 1958, a case that instantly assumed iconic status in the country’s medical culture. After pioneering organ transplantation in the 1970s, the Hospital became renowned in the 1990s
for breakthroughs in the management and molecular biology of leukemia. In terms of sheer volume, the
Hospital has around 60 masters’ and PhD students
at any one time, each with an accredited supervisor. Over 2 million patients annually attend its outpatient and emergency departments, and around
25 000 undergo surgery.
Hospital staff have an impressive array of over 300
awards to their credit, including the E. I. Evans Prize
(American Burns Association), the G. Whitaker International Burns Prize (Italy), Catherine Cancer
Prize (USA), the Charles Rodolphe Brupbacher
Foundation Prize (Switzerland), the Prix de l’Oise
(French Ligue Nationale Contre le Cancer), the SiProf Li Hongwei (李宏为). Prof Lin Yanzhen
mone
and Cino del Duca Foundation Prize (France),
Graduated from Shanghai (林言箴). Graduated
and the He Liang and He Li Fund (何梁何利基金)
Second Medical College in in 1949 from Aurora
1961. Tenured professor at
University. Tenured proTechnology Prize (Hong Kong).
Ruijin Hospital. Director
fessor at Ruijin Hospital.
In November 2002, France inaugurated its first
of the General Surgical
Current Honorary DirecSociety of the Chinese
large-scale cooperation structure, the Sino-France
tor of Shanghai Digestive
Medical Association. DiSurgery Institute and
Research Center for Life Science and Genome Rerector of the Department
Editor in Chief of the
search,
which was set up in Ruijin Hospital. This
of Surgery and Shanghai
Journal of Surgical
Digestive Surgical InstiTheories and Practice.
sealed 10 years’ collaboration between Ruijin, its
tute. Foreign member of
Foreign member of
Institute of Hematology under Dr Chen Zhu, and
French National Academy French National Academy
French
National Center for Scientific Research
of Surgery. Courtesy of
of Surgery. Courtesy of
Ruijin Hospital.
Ruijin Hospital.
(CNRS) laboratories UPR9051 and UMR7151 at
202 MEDICOGRAPHIA, VOL 31, No. 2, 2009
Ruijin Hospital – GU Qian, NI Lidong, and HU Weiguo
A TOUCH
Chinese Minister of Health
Chen Zhu (陈竺) speaking
during a press conference
on AIDS in China held
in Beijing, Thursday,
November 29, 2007.
© Ng Han Guan/AP/SIPA.
OF
FRANCE
Saint-Louis Hospital in Paris. An important research focus, headed by Chen Zhu, has been in acute promyelocytic leukemia, with particular regard to the mode of action of arsenic trioxide, the role of cyclic
adenosine monophosphate (cAMP), and the ubiquitination reaction. These and other studies have led to
publications in Science, Cancer Cell, Proceedings of the National Academy of Sciences, EMBO Journal,
Journal of Experimental Medicine, Journal of Clinical Investigation, Blood, American Journal of Human
Genetics, Oncogene, and Leukemia.
In 2004, Ruijin Hospital signed a cooperative agreement with Servier, the largest independent French
pharmaceutical company, to sponsor study visits to leading French hospitals by six Ruijin endocrinologists
and cardiologists. Servier has been present in China for over 20 years, during which time it has won the trust
of the country’s pharmacists and physicians. Over the last two decades, it has been working extensively
with Chinese academia via the Sino-French exchange program and cooperative research in epidemiology, public health, and other fields. Servier representatives regularly visit close on 1400 hospitals in China’s northern, southern, and eastern
provinces.
France has been unstinting in its recognition
of the outstanding contributions made by Ruijin
Hospital staff in fostering Sino-French partnerships. It has made Drs Kuang Ankun, Li Hongwei (李宏为), and Chen Zhu chevaliers of the Legion of Honor, while Drs Lin Yanzhen (林言箴),
Li Hongwei, and Chen Zhu have been elected
members of the French Academy of Sciences. In
many ways, Chen Zhu personifies the success of
Sino-French cooperation. After medical studies in Jiangxi, he took a postgraduate course at
Shanghai Second Medical University from where he went to Paris to earn a doctorate in hematology at
Saint-Louis Hospital. On 29 June 2007, he was appointed China’s Minister of Health, the second non–
Communist Party member—alongside Wan Gang (万钢) appointed Minister of Science and Technology
on 27 April 2007—to rise to such a key position since the late 1970s.
In June 2005, Ruijin Hospital and Shanghai Second Medical University hosted a week-long Sino-French
medical meeting cosponsored by the Chinese and French Ministries of Health. The sessions, largely devoted to HIV-AIDS (human immunodeficiency virus—acquired immunodeficiency syndrome), emergent
diseases, and the practicalities of medical exchange programs between the sponsor nations, were attended
by key figures in these areas, not least Chen Zhu, and, on the French side, Prof Vincent Deubel, director
of the Shanghai Pasteur Institute, and key advisers on health to the French government.
In 2007, Ruijin Hospital celebrated its 100th birthday in cultural continuity with the old Hôpital SainteMarie. French physicians were on hand to witness the unveiling of the centenary monument inscribed
with the hospital’s significant contributions (such as China’s first kidney, liver, and heart transplants), to
which their country had in no small measure contributed. It would have been clear to those present that
France had ceased to be the unchallenged mentor in the Sino-French medical relationship. Exchange was
no longer one-way. Increasing numbers of French physicians and bioscientists are now coming on training visits to Ruijin Hospital, to feed on the knowledge and experience of their Chinese colleagues, and to
bring back to France a love and appreciation for what this once-French, but now wholly Chinese hospital
is achieving in terms of both medical science and the care of its surrounding population. Ì
Cover of the Program
of the 27th FranceChina Medical Days
(27e Journées Médicales
France-Chine) in Beijing,
Wuhan, and Shanghai,
21-28 September 2008,
an illustration of one
among many longstanding partnerships between
China and Servier.
Ruijin Hospital – GU Qian, NI Lidong, and HU Weiguo
MEDICOGRAPHIA, VOL 31, No. 2, 2009 203
A TO
UCH
OF
FRA
NCE
L’HÔPITAL RUIJIN :
DE L’HÔPITAL FONDÉ PAR DES JÉSUITES FRANÇAIS AU XIXE SIÈCLE
À L’HÔPITAL UNIVERSITAIRE DE POINTE CHINOIS D’AUJOURD’HUI
’
L
Académie Aurore (Zhendan Xueyuan 震旦学院, mot à mot « le tonnerre du lever de soleil »,
est une institution catholique française fondée en 1903 à Shanghai par un Jésuite chinois, Joseph
Ma Xiangbo (马相伯), destinée à contribuer à la modernisation de la Chine. En 1907 la Société
de Jésus fit l’acquisition d’un terrain à proximité de l’Université Aurore sur lequel elle édifia l’Hôpital Sainte Marie, rebaptisé ultérieurement Hôpital Guangci (廣慈医院, mot à mot, « Hôpital
de la Grande Miséricorde »). Cet hôpital se transforma en centre d’enseignement pour les élèves
de l’Académie Aurore. En 1917 l’Académie fut promue au rang d’université, la prestigieuse Université
Aurore, et devint une pépinière de médecins, d’infirmières, et de techniciens médicaux pour les hôpitaux
catholiques de Chine. Une succession de doyens jésuites français en fit un phare de la médecine et de la
langue françaises où se côtoyaient étudiants français et chinois. En 1952, toutes les institutions éducatives étrangères en Chine passèrent sous direction chinoise et l’Université Aurore fusionna avec l’Université Fudan (复旦大学), tandis que son école de médecine fut intégrée à l’École de Médecine de l’Université de Shanghai No. 2. L’Hôpital Guangci, quant à lui, fut renommé Hôpital Ruijin (瑞金医院), pour
devenir un des centres hospitalo-universitaires de pointe de la Chine. Au cours des années 1980, l’École
de Médecine de l’Université de Shanghai No. 2 renoua avec son passé en proposant un enseignement de
7 ans de médecine en langue française et en établissant un programme très dynamique d’échanges entre
la Chine et la France à tous les niveaux. Nul n’illustre mieux le succès de cette entreprise que l’actuel Ministre de la santé chinoise, le Professeur Chen Zhu (陈竺), hématologue de renommée mondiale, dont le
brillant parcours médical s’effectua tant à l’École de Médecine de l’Université de Shanghai N° 2 qu’à la
Faculté de Médecine Paris VII. De nos jours, un nombre croissant de médecins et scientifiques français se
rendent à l’Hôpital Ruijin non seulement pour enseigner, mais pour apprendre. Les espoirs placés par le
fondateur jésuite Ma Xianbo dans son Académie se trouvent ainsi pleinement réalisés.
204 MEDICOGRAPHIA, VOL 31, No. 2, 2009
Ruijin Hospital – GU Qian, NI Lidong, and HU Weiguo
A
T
O U C H
O F
F
R A N C E
Dominique CAMUS, Journalist
6, rue du Porche
76590 Manéhouville, FRANCE
(e-mail: [email protected])
“Paris of the Orient”
The Shanghai French Concession
(1849-1946)
by D. Camus, France
T
he foreign concessions
It has often been said that the presence of the foreign nations who were granted settlements and
concessions in Shanghai by the 1842 Treaty of Nanjing contributed to jumpstart its transformation,
within less than a century, from a fishing village to one of Asia’s greatest metropolises. Yet with its
two to three hundred thousand inhabitants, mid-19th century Shanghai was already a thriving port,
whose junks sailed up and down the coast and across the East China Sea to Japan. However, the opening of the foreign concessions did prompt Chinese and Westerners alike to pursue a whole host of activities that propelled Shanghai into the 20th century.
Shanghai occupied a strategic position at the mouth of the Yangzi River, which the foreign nations were
quick to perceive as ideal for trade. In 1845, Intendant Gong, a well-read Confucian, drew up Land Regulations, which allocated the representatives of the French, British, and Americans agricultural land and
swamps north of the fortified town (several buildings of which still exist, even though drastically renovated,
in what is known as the “Old City,” surrounded by a circular avenue that has replaced the walls). In 1949,
an agreement was drafted by the Taotai (city official appointed by the Qing dynasty) Lin-Kuei, who allowed
the French to settle on land situated between the walls of the Chinese town and the Yangjingbang Creek.
S
hanghai, one of the world’s largest and most vibrant metropolises, with a population of over 20
million people, boasts a skyline that since the 1990s has undergone such dramatic changes that
frequent travelers to the city are constantly disoriented by the disappearance of old landmarks and
the practically overnight springing up of new skyscrapers vying with each other for world records
of height, avant-gardism, environmental sophistication, and sheer breathtaking beauty. Probably
the most symbolic example is the “Pudong New Area,” built on former farmland on the east side of
the Huangpu River, which has become China’s new financial hub, with its famous Shanghai World Financial Center (492 meters), Oriental Pearl Tower, with its 11 spheres, and Jin Mao building housing the
Grand Hyatt Hotel on the 36 uppermost of its 88 floors. Although sizeable areas of Old Shanghai have
been irretrievably lost in the process, much has been done to preserve major vestiges of the past. Most
conspicuous among the escapees of the wrecking ball are two historical districts that bear the architectural stamp of the foreign nations that were granted extraterritorial settlements and concessions in the
wake of the Nanjing Treaty, in 1842, which opened various ports, including that of Shanghai, to international trade. One is the “Bund,” which runs along one mile of the western bank of the Huangpu River and
is still lined by over 50 former British, Dutch, French, German, Japanese, Russian, and US banks, trading
houses, consulates, etc, of various styles. The other is the “French Concession,” where plane tree–lined
streets featuring many typically French mansions have been carefully preserved, and where a whiff of the
“Paris of the Orient,” as the district was called, still pervades the air.
www.medicographia.com
Medicographia. 2009;31:205-211.
“Paris of the Orient”: The Shanghai French Concession (1849-1946) – Camus
(see French abstract on page 211)
MEDICOGRAPHIA, VOL 31, No. 2, 2009 205
A T
OUCH
OF
FRANCE
View of Shanghai around
1860. Ink and watercolor on paper. Detail.
© Bridgeman Art Library.
During the turmoil of the Taiping Rebellion (1851-1864), notably when the Small Swords Society occupied the Chinese city in 1853, over 20 000 locals sought safety in the concessions, creating a social mix
that transformed the concessions into Sino-Western towns. One- or two-storey lilong houses (li means
communities and long signifies lanes, whence lane-and-community–based urban dwellings) were put up
everywhere and rented to local Chinese. Because of the increased demand for administrative services resulting from this sudden population upsurge, the British, French, and American consuls together drew up
municipal regulations in 1854, authorizing the foreign communities, represented by property owners, to
run the concessions. The resulting Shanghai Municipal Council fixed taxes, for foreigners and Chinese, to
fund urban projects and set up a police force. The French had misgivings about this creation of a single
administration, and in 1861 refused to ratify the new municipal regulations. The French Concession then
went it alone with an independent municipal council placed under the authority of the consul, in other
words, the Ministry of Foreign Affairs in Paris. In 1863, the British and American concessions fused and
took the name of the Shanghai International Settlement. This division of Shanghai and the concomitant
existence of rival authorities, jealously guarding their own prerogatives, contributed to the emergence of a
unique social and political space within the urban center destined to become the most influential metropolis of Asia in the years before World War II.
By the dawn of the 20th century, Western trading interests had spread beyond Shanghai through a
network of towns and ports around the coast and upcountry along the Yangzi River. Shanghai naturally
was the prime beneficiary of this growth. Through its port
passed over half of all Chinese imports and exports. Trading
firms and transport companies abounded and expansion in
international and waterborne trade opened up new markets. Driven mainly by private business, stores and artisans’
workshops sprang up across the city: the 1909 Shanghai
guide listed 430 shops; the number had risen to 520 in 1910,
and tripled by 1914.
The effectiveness of international trade was much dependent on rapid access to information. With the advent
of the telegraph (1865), information from London or Paris
could reach China in a few hours rather than thirty days,
and the Chinese quickly seized the opportunities offered by
this new means of communication.
The building of new roads to convey merchandise to
Shanghai’s warehouses also stimulated growth, as did the
rail link between Shanghai and Nanking (1902), which herAgreement instituting the French Concession,
alded
the building of tramways in the concessions and in
issued on 6 April 1849 by Lin-Kuei, the highthe Chinese town.
ranking Chinese official in charge of Shanghai
(Taotai), granting French Consul Charles de
The opening of the Suez Canal in 1869 shortened to two
Montigny territorial, building, and administramonths
or less the sea voyage to China, lent new impetus
tive rights. Diplomatic Archives of the French
Foreign Affairs Ministry. © Archives du MAE.
to the freight between Europe and Asia, and spurred devel-
206 MEDICOGRAPHIA, VOL 31, No. 2, 2009
“Paris of the Orient”: The Shanghai French Concession (1849-1946) – Camus
A TOUCH
OF
FRANCE
opment of newer steamships, since the narrow waterway was difficult for clippers to navigate. Shanghai
was upgraded to one of the world’s foremost ports thanks to dredging work by the Chinese administration
from 1906 to 1910, with the help of foreign engineers.
The French Concession
When he arrived in Shanghai in early 1848 as the first French Consul, Charles de Montigny’s mission was
to look after the interests of the French community, at this time little more than a handful of tradesmen
and missionaries. Anxious to fix the geographical limits of the French Concession, de Montigny negotiated an area of 66 hectares, bordered to the east by the Huangpu River, and to the north by the much larger
British Concession (199 hectares).
Public works
From the creation of the municipal council (1862), a series of public works took shape. One of the first
projects was the building of new roads, followed by work on drainage, paving, and sewers, funded by mandatory contributions from property owners. From 1887, the French Concession was the first to start a land
register in order to raise local and land taxes. Constructions were regulated and subject to approval by the
municipal council. Perspectives were defined, which doesn’t seem to have been the case for the International Settlement, whence the difficulty of joint realization of infrastructure work, such as the filling in
of the Yangjingbang Creek, which had become an open sewer. Common projects were, nonetheless, completed, such as the slaughterhouse, the cemetery, and a fire department water tank, and traffic and police
rules were decided jointly.
Created in 1906, the French Shanghai tram company fared well and by the 1930s had 100 trams, 60
buses, and 38 trolleybuses, and supplied countless households with electricity and water. Although its
salaries were somewhat lower than those of its rivals, the company was forward-looking and offered better welfare benefits: housing, relief funds, cost of hospitalization and treatment, school funds.
Map of Shanghai, dated 1931
showing the French Concession
(south), the International Settlement (north) resulting from
the merging of the British and
American Concessions. The
Chinese City is recognizable
by its circular boundary, still
marked today by the circular
road enclosing it, and the areas
in maroon north and east of
the Chinese city, labeled “Public
buildings, banks, and offices”
correspond to part of the Bund,
on the eastern bank of the
Huangpu (Whangpoo) River.
Map featured in “Mapping Urban
Infrastructure—Shanghai’s
New Concessions,” by B. Lai
and K. Tse. All rights reserved.
Banking
The arrival in Shanghai of the Comptoir National d’Escompte de Paris in 1860 launched French banking
in China. When it opened its Shanghai branch in 1898, the Banque de l’Indochine became the leading
French bank in China and led efforts to counter the hegemony of the British Hongkong and Shanghai
Banking Company Limited (HSBC), one of the oldest banking groups ever founded.
Back home in France, silk production could not meet the demand for luxury silk goods, long craved by
the French nobility and later by the middle classes that emerged during the industrial revolution. The
French State therefore decided to buy directly from China, and from 1848 to 1873 fifteen French silk merchants set up in Shanghai. Around 1900, Shanghai had sixteen silk mills, and the industry attracted more
foreign investors than any other.
Public health and welfare
Public health issues in Shanghai were addressed on the arrival of Westerners, no doubt impelled by the
dangers of unwholesome surroundings (swamps, canals) and a climate conducive to contagion. Improving public health was also a way for Westerners to win hearts and minds in the Chinese community. In 1844,
“Paris of the Orient”: The Shanghai French Concession (1849-1946) – Camus
MEDICOGRAPHIA, VOL 31, No. 2, 2009 207
A T
OUCH
OF
FRANCE
English missionaries set up the Shanghai Hospital, the first in the foreign concessions, and a few years
later French missionaries opened the General Shanghai Hospital and American missionaries the Hongkew
Hospital. As modern medicine was gradually put in place, health care establishments multiplied on the
initiative of foreign and Chinese doctors. Shanghai soon had the highest concentration of doctors in the
land, particularly those trained overseas (22%) or in foreign universities in China. In the 1930s, 1300 physicians trained in Western medicine and 5600 doctors of Chinese medicine tended the sick of Shanghai. No
other city in China could boast such a health care infrastructure.
French schools
The French Jesuits founded the Collège Saint Ignace, the first Catholic school in China, in 1847, and ten
years later it had 82 pupils, who studied Chinese characters, French, music, and drawing. Saint Ignatius
Cathedral was built between 1905 and 1910. Children of Christian families received both a religious education and literary training to enable them to sit the exams for the Chinese baccalaureate. In 1886, the
French municipality created and financed the
Franco-Chinese municipal school, the first
of its kind. Teaching was entrusted to the Jesuits and then to the Marist Brothers. Young
Chinese from the working classes received
instruction and often went on to work in
business, the administration, or the railways.
Chinese pupils on scholarships were sent to
France to achieve mastery of French language
and culture so that they would create businesses on their return to their homeland. In
addition to the cathedral, the French Jesuits
also built orphanages, monasteries, schools,
libraries, and an observatory.
Aurore University
Ma Xiangbo, a Jesuit catholic priest and man
of letters, founded Aurore University, the jewInterior of Saint Ignatius-Xujiahui Cathedral, the largest Catholic cathedral in the Far
el of French educational establishments in
East at the time of its construction in 1905-1910 in the French Concession. The Church
China, in 1902. This Franco-Chinese instituis being restored for the Shanghai Expo 2010 World Fair. © AKG-Images/Bruce Conolly.
tion included departments of literature, philosophy, mathematics, and natural sciences, to which were added in 1914 faculties of arts-law, civil engineering, and medicine. Upon completion of a six-year course, Aurore medical graduates were eligible
to enter the fifth year of medical school at Paris University. After a civil engineering degree (five years),
awarded on presentation of a technical project, a graduate could enter the National College of Electrical
Engineering in Paris.
Following differences of opinion with the French Fathers, Ma Xiangbo in 1905 created Fudan University, whose diplomas were recognized by both the French and Chinese governments, and which admitted
its first female students in 1937.
The arts
The emergence of a Chinese bourgeoisie in Shanghai led to the creation of a school of guohua painting
that produced great painters and calligraphers at the end of the Qing Dynasty (1644-1912) and during
the Republic (1912-1949). The Jesuits at Zikawei ran an orphanage, a printing works, the site of China’s
first lithographic press (1876), and a center of Western arts and crafts, where Chinese artists could take
classes in drawing and anatomy, thus breaking with their artistic tradition. Zhou Xiang, a functionary
of the tenth Qing Emperor, who had spent several years in France, established a school that pioneered
Occidentalism in Shanghai, particularly in the illustrated press. In 1912 one of its students, Liu Haisu,
founded in the French Concession the veritable forerunner of modern schools of fine arts, a mixed academy whose revolutionary teaching methods drew inspiration from those of the Parisian academies, where
thirty or more future Chinese masters studied fine arts between 1910 and 1930. Sartorial inspiration too
was to be seen in the concession’s “little Montmartre” where, after the manner of French artists, floppy
neckties were all the rage.
Lin Fengmian, studied in Dijon, Paris, and Berlin from 1918 to 1926, married the French sculptress
Alice Vattant, and on his return to China was appointed director of the Peking Institute of the Arts. Later
he ran the new National Academy of Art in Hangzhou, near Shanghai, where, influenced by impression-
208 MEDICOGRAPHIA, VOL 31, No. 2, 2009
“Paris of the Orient”: The Shanghai French Concession (1849-1946) – Camus
A TOUCH
OF
FRANCE
ism, he taught oil painting. This prestigious school became the
center of Chinese modernism from which other remarkable painters were later to emerge, notably the Frenchmen Zao Wou-ki (who
exhibited in 2008 at the Bibliothèque Nationale de Paris) and
Chu Teh-Chun, and the Americans Chao Chung-Hsiang and Wu
Guanzhong.
Pang Xunqin, who studied medicine at Aurore University and
music in France, presented his canvasses at the 1925 Paris Exhibition. On his return to Shanghai in 1930, he created the Société des
Deux Mondes, later known as “The Storm Society,” which organized four milestone exhibitions in the French Concession. André
Breton’s Surrealist Manifesto was translated into Chinese in 1935
on the occasion of the first surrealist exhibition organized by the
French Concession for Independent Chinese Artists.
Father Chevalier in
1919 at Zikawei
(Xujiahui) Observatory,
founded by the French
Jesuits in 1872.
© Ed. Belin, Paris.
Architectural styles
In the 1920s, along the Bund, the urban waterfront on the Huangpu
River, foreign wealth and power were incarnate in elegant, even
vainglorious, monumental buildings. This craze for ever higher
edifices was not shared by the French, whose architectural legacy
can be seen to this day in fine buildings like the Chung Wai Bank, as well as the Gascogne, the Béarn,
and the Picardie, all three classified as historic buildings by the Shanghai Municipality, whose Art Deco
charm, like that of the Normandie (1937-1938), has not faded with the passing of the years.
Cultural life and leisure activities
The foreign presence in Shanghai galvanized the newspaper business and publishing. First published in
1850, the British weekly North China Herald included a supplement in Chinese from 1861, was renamed
three years later as the North China Daily News, and became the most influential foreign newspaper of
its time. Le Journal de Shanghai catered for the French community, and Shen Bao, first published in 1872
and written entirely in Chinese, was soon the most powerful newspaper in China and remained so until
1949. In the concessions there were 2487 bookstores in 1938, compared with 136 in Peking and 40 in
Nanking. It therefore comes as no surprise to learn that between 1920 and 1945 Shanghai counted the
largest number of intellectuals of any city in China.
Shanghai had long enjoyed a nightlife worthy of a port and commercial hub, and by the 1920s its range
of distractions was unrivaled in China. From 1912, the Grand Monde, one of the main centers of entertainment, hosted a panoply of distractions— theater, cafés, restaurants, gaming rooms, and cinema (by
1940 there were 57 film theaters in Shanghai). Gambling, one of the great Chinese passions, flourished in
the concessions despite its prohibition under the Qing Dynasty and then under the Republic. The social life
of Westerners in the concessions centered on their clubs. Each nationality had its own and the archetype
A view of the famous, still extant Bund (Huangpo River Embankment) and
its European-style buildings. © Bettmann/Corbis.
“Paris of the Orient”: The Shanghai French Concession (1849-1946) – Camus
Leafy archway of plane trees in today’s French Concession in Shanghai.
“Quiet Streets,” photo by Alan Levine. With kind permission.
MEDICOGRAPHIA, VOL 31, No. 2, 2009 209
A T
OUCH
OF
FRANCE
was the British residents’ Shanghai Club, famous for its
marbles, paneling, and bar of over 30 meters—the “longest
in the world.” The price of admission though was beyond
the means of ordinary residents. The French Sports Club,
with its fine ballroom and covered swimming pool, was the
most popular venue.
Normandy-style cottage
in the French Concession
now housing the
Shanghai Institute for
Advanced Studies (SIAS).
Photo by Eecc. With
kind permission.
Crescent-roofed
Shanghai Grand Theater in
People’s Square, designed
by French architect
Jean-Marie Charpentier.
© Jon Hicks/Corbis.
Concourse windows
at Pudong International
Airport, designed by French
architect Paul Andreu.
© Yang Liu/Corbis.
The curtain falls… and rises again
In the 1940s, after close to a century, the end of the concessions drew near. When the Imperial Japanese Army occupied Shanghai in 1941, hundreds of thousands of displaced
Chinese poured into the concessions, which became foreign
islands in a sea of invading Japanese forces. The Sino-British
Friendship Treaty of February 1943 returned the International Settlement to Chinese control, and on 30 July 1943 the
Consul General Roland de Margerie officially handed over
the keys of the French Concession to the Mayor of Shanghai.
Another 50 years later, what is left of the French Concession? Has anything survived the wrecking ball?
The first thing that jumps to the eye is the plane trees that liberally line the streets and avenues of the
former French district. This “greenery” in the modern city of steel, concrete, and glass is a welcome reminder of less hectic days and of the French way of life. Ironically what the French planted were “London
planes,” but these trees immediately took the fancy of the Chinese who rechristened them “French planes.”
The French Concession remained largely untouched until the 1980s, when Shanghai started its drive to
develop into the megalopolis of today.
French names of streets have given way to Chinese names, many buildings have been torn down, and
even the trees planted by the French on the former Avenue Joffre were removed—only for new trees to be
hastily planted back to placate an angry neighborhood population. Plane trees have become so popular
that they spill out in Shanghai beyond the bounds of the former French Concession and are even now
found as roadside trees throughout China. Since the 2000s, the municipality of Shanghai has been taking
active measures to preserve as much as possible of the vestiges of the French presence and is restoring old
buildings and even some of the “lilongs” of the French Concession. The French Concession is, today, one
of Shanghai’s major tourist attractions and the atmosphere of the “Paris of the Orient” is still palpable.
But the story doesn’t stop there, and the “Touch of France” (to allude to the title of the cultural section
of this journal) has come back in the form of
a spate of French architects who have been or
are being commissioned by the municipality
of present-day Shanghai to leave their imprint
on the city.
Jean-Marie Charpentier designed the Shanghai Grand Theater (“Opera”) situated near People’s Square in 1998; he landscaped a section
of Nanjing Avenue (Nanjing Donglu) in 1999,
which has become Shanghai’s bustling main
pedestrian restaurant and shopping avenue, as
well as the 4.5-km Century Avenue, the major
thoroughfare destined to structure the 520 km2
of the financial and commercial hub of Pudong
and become Shanghai’s “Champs-Élysées”
(2000); he built the General Motors Headquarters on the same Century Avenue (2004); the
Shanghai Entry and Exit Visa Administration
Bureau in Pudong (2005); and the Saint-Gobain
(glass manufacturer) R&D Center, in the Minhang Development Zone (2007). Jean-Marie
Charpentier has also been commissioned to
rehabilitate an area of 50 000 m2 in the French
Concession to be completed in 2010. Paul Andreu designed Shanghai International Airport
in 1999; the Oriental Arts Center in Pudong, in
210 MEDICOGRAPHIA, VOL 31, No. 2, 2009
“Paris of the Orient”: The Shanghai French Concession (1849-1946) – Camus
A TOUCH
OF
FRANCE
2004 (as well as the National Opera House in Tiananmen Square, Beijing in 2006). Jean-Marie Duthilleul
designed the Shanghai South Railway Station in 2006. And to end this (nonexhaustive) list of major projects, Antoine Grumbach was commissioned in February 2008 to renovate 150 000 m2 of Shanghai’s 19thcentury historic center, on time for the Shanghai Expo 2010 World Fair.
Thus the French presence in Shanghai is not only a nostalgic thing of the past, but is even today actively
contributing to shape the world’s most superlative city. FURTHER READING
– Bergère MC. Histoire de Shanghai. Paris, France: Fayard; 2002.
– La France en Chine (1843-1943). Nantes, France: Archives diplomatiques.
– Henriot C, Roux A. Le Shanghai des années 30, plaisirs et violences. Paris, France: L’Harmattan; 1998.
– Le Paris de l’Orient (1849-1846), Présence Française à Shan-
ghai, catalogue de l’exhibition musée Albert Khan, 2002.
– Brossollet G. Les Français de Shanghai. Paris, France: Belin;
1999.
– Henriot C. Atlas de Shanghai. France CNRS; 1999.
– Roux A. Le Shanghai Ouvrier des Années 30. Paris, France:
L’Harmattan; 1993.
From fishing village to
vibrant megalopolis: impressionistic sunrise in Pudong,
Shanghai, that could have
been painted by Monet.
© Paul Hardy, Corbis.
LA
“LE PARIS DE L’ORIENT” :
CONCESSION FRANÇAISE DE SHANGHAI (1849-1946)
S
hanghai, une des métropoles les plus gigantesques et animées du monde avec ses plus de 20
millions d’habitants, a subi depuis 1990 des changements si spectaculaires que les voyageurs
s’y rendant fréquemment sont à chaque fois désorientés par la disparition de leurs points de repère et l’apparition du jour au lendemain de nouveaux gratte-ciel rivalisant les uns avec les
autres pour établir des records mondiaux de hauteur, d’avant-gardisme, de sophistication environnementale et de beauté à couper le souffle. L’exemple probablement le plus symbolique est celui de la zone nouvelle de Pudong, construite sur d’anciennes terres cultivées de la rive orientale de la
rivière Huangpu, et devenue le nouveau cœur financier chinois, avec son célèbre « Shanghai World Financial Center » (492 m), la Perle de l’Orient avec ses 11 sphères et la tour Jin Mao accueillant l’hôtel Grand Hyatt aux 36 derniers de ses 88 étages. Bien que de vastes étendues du Vieux Shanghai aient
irrémédiablement disparu au cours des réaménagements urbains successifs, nombre de vestiges du
passé ont pu être sauvegardés. Ont ainsi échappé au rouleau compresseur des démolitions deux quartiers historiques portant l’empreinte architecturale des nations étrangères à qui le traité de Nanjing de
1842, qui ouvrit plusieurs ports au commerce international dont Shanghai, octroya des enclaves territoriales, les « concessions ». L’un de ces quartiers est le « Bund », qui s’étend sur un kilomètre sur la
rive occidentale de la rivière Huangpu et qui est toujours jalonné de plus de 50 banques, sièges de sociétés, consulats, etc., allemandes, américaines, anglaises, françaises, japonaises, néerlandaises et
russes, aux styles les plus variés. L’autre est la « Concession Française », où de nombreuses habitations
typiquement françaises soigneusement préservées s’élèvent au milieu de rues bordées de platanes et où
flotte encore une atmosphère du « Paris de l’Orient »d’autrefois.
“Paris of the Orient”: The Shanghai French Concession (1849-1946) – Camus
MEDICOGRAPHIA, VOL 31, No. 2, 2009 211
Medicographia
A
Ser vier
publication
I nstructions
for authors
General instructions
N Manuscripts should be provided by e-mail ([email protected].
com) or by CD double-spaced, with 2.5-cm margins. Pages must
be numbered. Standard typed page = 25 lines of 90 characters
(including spaces) double-spaced, 2.5-cm margins = a total of
about 320 words per page.
N All texts should be submitted in English.
N Provide 1 color photograph of main author.
N On the title page, provide: a title (concise and informative); full
names of authors (first name, middle name initial, and last name);
highest academic degrees (in country-of-origin language); affiliations (names of department[s] and institution[s] at the time the work
was done); a short running title (no more than 50 letters and spaces),
keywords (5-10); corresponding author’s complete mailing address
and telephone No., fax No., and e-mail address; acknowledgments
(on title page, or at end of main text).
N Include an Abstract of 200-230 words for all texts except Editorials and replies to the Controversial Question.
N Figures and Tables. Figures should be of good quality or professionally prepared, numbered according to their order, with proper
orientation indicated (eg, "top," or "left"). Figures may be provided
as pdf files (printing resolution = 300 dpi scans, on CDrom, or via
e-mail; screen resolution = 72 dpi scans acceptable only if largesized format [A4]). Provide fully explicit legends, not repetitive of
text. All abbreviations used should be explained in the legends. As
figures and graphs may need to be reduced or enlarged, all absolute
values and statistics should be provided. Illustrations will be reproduced in full color only when clearly necessary, eg, images from nuclear medicine or histology. Provide each table on a separate sheet,
with title above and description below. All figures and tables should
be cited in the text, with distinct numbering for figures and tables.
N Note that Editorials and Abstracts will be published in English
and French. Translations into French will be provided by the Publisher’s Editorial Department.
N Include Headings using a consistent style for the various levels
of headings, to highlight key points and facilitate comprehension of
the text. The Editorial Department reserves the right to add or delete
headings when necessary.
N Abbreviations should be used sparingly and expanded at first
mention. A list of selected abbreviations and acronyms should be
provided (or will be prepared by the Editorial Department) where
necessary.
N Use Système International (SI) units.
N Use generic names of drugs.
N All references should be cited in the text and numbered consecutively using superscript arabic numerals. Presentation of the
references should be based on the Uniform Requirements for Manuscripts Submitted to Biomedical Journals. Ann Intern Med. 1997;
126:36-47 (“Vancouver style”). The author-date system of citation
is NOT acceptable. In press references are to be avoided. In the
bibliography, titles of journals should be abbreviated according to
the Index Medicus. All authors should be listed up to six; if there
are more, only the first three should be listed, followed by “et al.”
Where necessary, references will be styled by the Editorial Department to Medicographia copyediting requirements. Authors bear
total responsibility for the accuracy and completeness of all references and for correct text citation. Example of style for references:
1. Ouriel K, Geary K, Green RM, Geary JE, DeWeese JA. Factors
determining survival after ruptured abdominal aneurysm. J Vasc Surg.
1990;11:493-496.
2. Darling RC, Brewster DC, Ottinger LW. Autopsy study of unoperated abdominal aortic aneurysms: the case for early resection.
Circulation. 1977;56(suppl II):II161-II164.
3. Schulman JL. Immunology of influenza. In: Kilbourne ED, Alfade
RT, eds. The Influenza Viruses and Influenza. Orlando, Fla: Academic
Press Inc; 1975:373-393.
Specific formats
N Editorial: 1500 words. No abstract or illustrations should be includ-
ed. A French translation of the Editorial will be provided by the Editorial Department and submitted to the author.
N Theme - Focus - Update - Therapeutic outlook article - Touch
of France: Abstract: 200-230 words. Main text: 2800-3200 words.
References: their number should not exceed 50. Illustrations (figures and tables): their number should not exceed 5 unless clearly
necessary.
N Interview: Abstract: 200-230 words. Main text: 2000-2500 words.
Headings are the questions posed at the interview. References, if
cited, should in no case exceed 10. No illustrations.
N Replies to the Controversial Question: 400-600 words. No abstract or illustrations should be included. References, if cited, should
in no case exceed 6.
Editorial processing
N Editorial style: All contributions to Medicographia will be styled
by the Editorial Department according to the specifications of the current edition of the American Medical Association Manual of Style,
Williams & Wilkins.
N Page proofs and editorial queries will be sent to the corresponding author for approval. Corrections should be returned within 48
hours by e-mail, and fax or express mail. If this deadline is not met,
changes made by the Editorial Department will be assumed to be
accepted by the author. Authors are responsible for all statements
made in their work, including changes made by the Editorial Department and authorized by the author. Articles and abstracts will be edited to required length or returned to the author if specific requirements
are not complied with.
Copyright
N Copyright of articles will be transferred to the Publisher of Medicographia. The Copyright Transfer Agreement must be signed by the
main author and all coauthors and returned to the Publisher.
N For reproduction of copyrighted work, it is the author’s responsibility to obtain authorizations from the author(s) (including self) and
the publisher(s) and provide copies of these authorizations with the
manuscript.