use of ketamine as a fast acting antidepressant for

112
USE OF KETAMINE AS A FAST ACTING ANTIDEPRESSANT FOR TERMINAL CANCER PATIENTS WITH MAJOR DEPRESSIVE DISORDER Claudia Grott Zanicotti A thesis submitted for the degree of Master of Medical Sciences of the University of Otago Dunedin School of Medicine New Zealand March 2012

Upload: others

Post on 03-Oct-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

USE OF KETAMINE AS A FAST ACTING

ANTIDEPRESSANT FOR TERMINAL CANCER

PATIENTS WITH MAJOR DEPRESSIVE

DISORDER

Claudia Grott Zanicotti

A thesis submitted for the degree of Master of Medical Sciences

of the University of Otago

Dunedin School of Medicine

New Zealand

March 2012

! """!

ACKNOWLEDGEMENTS

After one year of new experiences and a complete change in my lifestyle, I can look back and

see how much I have learned. Obviously, the certainty that there is much more knowledge

ahead is a factor that keeps me motivated, but without the support from my loved friends and

family I would have never achieved one more goal. Thank you all!

It has been a hard year for my family, as we are physically very distant, but emotionally

incredibly close. I hope to see you soon (in New Zealand). I would also like to thank my

husband for being so supportive at all times, I would not be here without you. I love you all!

I would like to thank the staff from the Oncology Department, specially the oncology nurses

and receptionists. Your help with patient’s charts and arranging appointments was essential in

my learning process. I’m also grateful to the Psychological Medicine Department staff,

particularly for been so helpful and welcoming. Roz and Jill: thank you for all your help, for

the arrangements with conferences, and for always clarifying any doubts or questions. I was

also lucky to meet some great people during my stay in the first floor and I really enjoyed our

brief but unforgettable talks, and I sincerely hope these were just the first of many. Sarah was

the best office mate I could have ever found, incredibly friendly and respectful. Also, I am

thankful for the support from Otago Medical Research Foundation Inc. and the Otago

Community Trust.

I am thankful for the help and support of my supervisor and co-supervisor, Professor Paul

Glue and Associate Professor David Perez. Your constant feedback kept me positive during

the toughest times. Professor Paul, I am eternally grateful for your support since the very

beginning, even before I came to New Zealand. Thank you for your constant patience and for

dedicating so much time to this thesis. I definitely met an amazing professional and a person

of incredible character.

My very special thanks goes to the patient who made all of this possible. You are a unique

person, and your patience with the constant assessments was very kind of you. Thank you so

much!

! "#!

ABSTRACT

The objective of this thesis was to investigate the safety and efficacy of single and repeated

administration of ketamine as a fast acting antidepressant in patients with terminal cancer.

Associated with this objective was a comprehensive review of the pathophysiology of major

depressive disorder, existing treatments for major depressive disorder, and a review and meta-

analysis of treatments for depression in patients with cancer. The use of ketamine in

treatment-resistant depression was also reviewed, along with a meta-analysis of mood

responses in placebo-controlled studies, and data from case reports and case series.

The review and meta-analysis of antidepressant treatments for depression in cancer patients

yielded only two small placebo-controlled randomized trials, both for mianserin. No

psychotherapy trials were found that met inclusion criteria for this analysis. The two

mianserin trials showed a benefit in improving mood ratings compared with placebo. The

very limited amount of data on treatment of depression in this population is surprising, and

further research is needed to improve knowledge in this area.

The review and meta-analysis of mood responses to ketamine in three placebo-controlled

randomized crossover studies demonstrated a robust improvement in mood ratings by four

hours, that was sustained up to 72 hours. Uncontrolled data from case series and case reports

showed similar profile to data reported from the placebo-controlled studies.

The study investigates the effect of ketamine on mood ratings in depressed patients with

cancer had three stages; (1) to assess whether a single 1 mg/kg IM dose improved mood; (2)

to assess the effect of repeated 1 mg/kg IM dosing on mood; and (3) to assess the mood

responses to IM doses of 0.1, 0.5 or 1.0 mg/kg IM. This thesis reports the case of a single

patient who participated in Stages 1 and 2. The results in the case presented in this thesis were

positive, with remission of symptoms with single dose and repeated treatment, and

maintenance of remission with weekly injections of ketamine 1 mg/kg.

This thesis supports the possible use of ketamine as a fast acting antidepressant in terminal

cancer patients with Major Depressive Disorder. In a population with short life expectancy,

the existence of a treatment for depressive episodes with rapid onset of action would be

essential, and ketamine might be the option of choice.

! #!

TABLE OF CONTENTS

AUTHOR DECLARATION ........................................................................................... ii

ACKNOWLEDGEMENTS ............................................................................................ iii

ABSTRACT ...................................................................................................................... iv

TABLE OF CONTENTS .................................................................................................. v

LIST OF PUBLICATIONS ............................................................................................ ix

LIST OF FIGURES .......................................................................................................... x

LIST OF TABLES .......................................................................................................... xii

LIST OF ABBREVIATIONS ....................................................................................... xiii

1 INTRODUCTION ............................................................................................... 16

1.1 Introduction ......................................................................................................... 17

1.2 History of depression .......................................................................................... 18

1.3 Epidemiology of depression in Major Depressive Disorder and Bipolar

Disorder ………………………………………...………………………………. 19

1.4 Epidemiology of Major Depressive Disorder in cancer patients .................... 21

2 PATHOPHYSIOLOGY AND TREATMENT OF MAJOR DEPRESSIVE

DISORDER ……………………………………………………..……………… 23

2.1 Pathophysiology of Major Depressive Disorder ............................................... 24

2.1.1 Serotonergic system .............................................................................................. 26

2.1.2 Noradrenergic system ........................................................................................... 26

2.1.3 Dopaminergic system ............................................................................................ 27

2.1.4 HPA axis ............................................................................................................... 27

2.1.5 Gabaergic system .................................................................................................. 29

! #"!

2.1.6 Glutamatergic system ............................................................................................ 29

2.1.7 Cytokines .............................................................................................................. 31

2.2 Current treatments for depressive episodes ..................................................... 32

2.2.1 Psychotherapy ....................................................................................................... 32

2.2.2 Pharmacotherapy .................................................................................................... 32

2.2.3 Electroconvulsive therapy ..................................................................................... 33

2.2.4 Repetitive transcranial magnetic stimulation ........................................................ 34

2.2.5 Vagus nerve stimulation ........................................................................................ 34

2.2.6 Deep brain stimulation .......................................................................................... 35

3 REVIEW AND META-ANALYSIS OF AVAILABLE

TREATMENTS FOR DEPRESSION IN CANCER PATIENTS .................. 36

3.1 Introduction …………………………………………………………………… 37

3.2 Methods ………………………………………………………………….…….. 37

3.2.1 Search strategy ……………………………………………………...…...……… 37

3.2.2 Selection criteria …………………………………………..…………....………. 37

3.2.3 Analysis …………………………………………………..…………...………… 39

3.3 Results …………………….……………………………………………………. 39

3.3.1 Meta-analysis ……………….…………………………………………..……… 39

3.4 Discussion ………………………………………………………………………. 40

3.5 Conclusions …………………………………………………………………….. 41

4 KETAMINE OVERVIEW ………………………………………………...….. 43

4.1 Ketamine .............................................................................................................. 44

4.2 Ketamine as an anesthetic .................................................................................. 44

4.3 Ketamine as a drug of abuse .............................................................................. 44

! #""!

4.4 Ketamine as an analgesic .................................................................................... 45

4.5 Ketamine as a fast acting antidepressant ……………………..……………… 45

4.6 The possible use of ketamine as a fast acting antidepressant for terminal

cancer patients .................................................................................................... 46

5 REVIEW AND META-ANALYSIS OF KETAMINE AS A FAST ACTING

ANTIDEPRESSANT .......................................................................................... 47

5.1 Introduction ......................................................................................................... 48

5.2 Methods ................................................................................................................ 49

5.2.1 Search strategy ...................................................................................................... 49

5.2.2 Selection criteria ................................................................................................... 49

5.2.3 Analyses ................................................................................................................ 51

5.3 Results .................................................................................................................. 51

5.3.1 Randomized clinical trials ..................................................................................... 51

5.3.2 Case reports and case series .................................................................................. 54

5.3.3 Meta-analysis ........................................................................................................ 59

5.4 Discussion ............................................................................................................. 61

5.5 Conclusions .......................................................................................................... 62

6 PROTOCOL FOR TESTING KETAMINE IN MAJOR DEPRESSIVE

DISORDER IN TERMINAL CANCER PATIENTS ...................................... 63

6.1 Introduction ......................................................................................................... 64

6.2 Stages .................................................................................................................... 64

6.2.1 Stage 1 ................................................................................................................... 64

6.2.2 Stage 2 ................................................................................................................... 64

6.2.3 Stage 3 ................................................................................................................... 65

! #"""!

6.3 Inclusion and exclusion criteria ......................................................................... 65

6.3.1 Inclusion criteria ................................................................................................... 65

6.3.2 Exclusion criteria .................................................................................................. 65

6.4 Informed consent ................................................................................................ 69

6.5 Assessments ......................................................................................................... 69

6.6 Statistical analysis ………………………………………………….………….. 72

6.7 Conclusions ……………………………………………………….…………… 72

7 ACUTE AND MAINTENANCE IM KETAMINE FOR A DEPRESSED

PATIENT WITH TERMINAL CANCER ....................................................... 73

7.1 Introduction ......................................................................................................... 74

7.2 Case report ........................................................................................................... 74

7.3 Discussion ............................................................................................................. 81

7.4 Conclusions .......................................................................................................... 82

8 CONCLUSIONS ................................................................................................. 83

8.1 Conclusions …………………………………………………………….………. 84

9 REFERENCES .................................................................................................... 85

9.1 References ……………………………………………………………………… 86

APPENDIXES

Appendix I- Informed consent 103

Appendix II- Case report - in press (page proofs) 109

! "$!

LIST OF PUBLICATIONS

Zanicotti CG, Perez D, Glue P. Acute and maintenance IM ketamine for a depressed patient

with terminal cancer. Oral presentation at: The Royal Australian and New Zealand College of

Psychiatrists (RANZCP) 2011 New Zealand Conference. 2011 Sep 28-30; Queenstown, New

Zealand.

Zanicotti CG, Perez D, Glue P. Effects of low dose intramuscular ketamine on depression

ratings in depressed patients with cancer in palliative care – a case report (Efeitos da

quetamina via intramuscular em baixa dose nos escores de depressão em pacientes deprimidos

e com cancer sob cuidados paliativos – relato de caso). Oral presentation at: 11º Congresso

Brasileiro de Clínica Médica (SBCM). 2011 Oct 27-29. Curitiba, Brazil.

Zanicotti CG, Perez D, Glue P: Mood and pain responses to repeat doses intramuscular

ketamine in a depressed patient with advanced cancer. J Palliat Med 2011 [in press].

! $!

LIST OF FIGURES

Figure 2.1: Anatomical connections and interactions between neuroendocrine system,

BDNF, monoamines, and cytokines in the brain.

Figure 2.2: HPA axis dysfunction in MDD.

Figure 2.3: Tripartide glutamatergic synapse.

Figure 2.4: From inflammation to depressive symptoms.

Figure 3.1: Selection of trials for meta-analysis.

Figure 3.2: Meta-analysis of antidepressant trials in patients with MDD and cancer.

Figure 4.1: Relation between ketamine dosing and its indications.

Figure 5.1: Selection of studies for review and meta-analysis for ketamine as a fast acting

antidepressant.

Figure 5.2: HAM-D mean values for Diazgranados et al., 2010, Zarate et al., 2006, and

Berman et al., 2000.

Figure 5.3: Cases outcomes and Diazgranados et al., 2010 (HAMD).

! $"!

Figure 5.4: Cases outcomes and Diazgranados et al., 2010 (MADRS).

Figure 5.5: Case outcome and Diazgranados et al., 2010 (BDI).

Figure 5.6: Meta-analysis results based on change in HAMD scores.

Figure 6.1: MADRS.

Figure 6.2: HADS.

Figure 6.3: The Demoralization Scale.

Figure 7.1: Antidepressant and analgesic response to IM ketamine 1 mg/kg.

Figure 7.2: Changes in HADS and Demoralization scales with IM ketamine 1 mg/kg.

Figure 7.3: Pearson’s correlation between change in depression and pain scores.

Figure 7.4: Antidepressant response to repeated dosing, at intervals of 7 days and > 7 days.

! $""!

LIST OF TABLES

Table 2.1: Main mechanisms of action of antidepressants.

Table 5.1: Demographic and clinical data for RCT’s.

Table 5.2: Demographic and clinical data for cases.

Table 6.1: ECOG performance status.

Table 6.2: The Comparative Pain Scale.

! $"""!

LIST OF ABBREVIATIONS

ACTH Adrenocorticotropic hormone

ACTRN Australian New Zealand Clinical Trials Registry

AD Alzheimer’s disease

AMPA Alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid

AMPARs AMPA receptors

BD Bipolar disorder

BDI Beck Depression Inventory

BDNF Brain-derived neurotrophic factor

CMI Cell-mediated immune

COPD Chronic obstructive pulmonary disease

CRF Corticotropin-releasing factor

CRH Corticotropin-releasing hormone

CVD Cardiovascular disorder

DBS Deep brain stimulation

DPP IV Dipeptidyl peptidase IV

DS Demoralization scale

DSM Diagnostic and Statistical Manual of Mental Disorders

EAAT Excitatory amino-acid transporter

EC Endocannabinoid

ECOG Eastern Cooperative Oncology Group

ECT Electroconvulsive therapy

GABA Gamma-aminobutyric acid

! $"#!

Gln Glutamine

Glu Glutamate

GluTs Vesicular glutamate transporters

HADS Hospital Anxiety and Depression Scale

HAMD Hamilton Rating Scale for Depression

HD Huntington disease

HIV Human immunodeficiency virus

HPA Hypothalamo-Pituitary-Adrenal

5-HT 5-hydroxytryptamine (serotonin)

HVA Homovanilic acid

IBD Inflammatory bowel disease

ICD International Classification of Diseases

IDO Indoleamine-2,3-dioxygenase

IFN Interferon

IL Interleukin

IM Intramuscular

IV Intravenous

LC Locus coeruleus

MADRS Montgomery-Asberg Depression Rating Scale

MAOI Monoamine oxidase inhibitor

MDD Major Depressive Disorder

MDE Major depressive episode

mGlu Metabotropic glutamate

mGluR Metabotropic glutamate receptor

! $#!

MRS Magnetic Resonance Spectroscopy

MS Multiple sclerosis

mTOR Mammalian target of paramycin

NA Nucleus accumbens

NMDA N-methyl-D-aspartate

NMDARs NMDA receptors

PD Parkinson’s disease

PEP Prolyl endopeptidase

PFC Prefrontal cortex

QIDS-SR 16 The Quick Inventory of Depressive Symptomatology –Self Report (16

items)

RCT Randomized clinical trials

SD Standard deviation

SERT 5-HT transporter

SLE Systemic lupus erythematosus

SNARE Soluble N-ethylmaleimide sensitive factor attachment protein receptor

SNRI Serotonin-norepinephrine reuptake inhibitors

SSRI Selective serotonina reuptake inhibitors

TMS Transcranial magnetic stimulation

TNF Tumor necrosis factor

TRYCATs Tryptophan catabolites

VNS Vagus nerve stimulation

VTA Ventral tegmental area

WHO World health Organization

! %&!

CHAPTER 1

Introduction

!

! %'!

1.1 Introduction!Major Depressive Disorder (MDD) is the most common psychiatric illness, and it affects

more than 120 million people worldwide (more than 27 times the population of New

Zealand). It is characterized by depressed mood and loss of interest and pleasure, but other

signs and symptoms are commonly present, such as: weight loss or gain; sleep disturbance

with insomnia or hypersomnia; fatigue; lack of concentration; feelings of guilt; suicidal

ideation; and psychomotor agitation or retardation (Skolnick et al., 2009; Statistics New

Zealand, Jan 2012; DSM IV). These symptoms must persist for at least 2 weeks, and must be

accompanied by significant distress or impairment. Also, in addition to its high prevalence,

depression presents the greatest proportion of burden due to nonfatal health outcomes (World

Health Organization - World Health Statistics, 2007). !!In patients with cancer, the prevalence of depressive episodes is higher than in general

population (Mitchell et al., 2011). Although it may seem unsurprising, special attention must

be given to this group, as there is a higher risk for depressed patients not to accept proper

cancer treatment (Colleoni et al., 2000). Thus, any depressive symptom should be recorded

and questioned in every follow-up appointment, and adequate treatment should be provided as

fast as possible, in the presence of a depressive episode. Available treatments for depression

will be described further in this chapter.!!Ketamine is an anaesthetic agent that has demonstrated antidepressant properties

(Diazgranados et al.,2010; Berman et al., 2000; Sofia and Harakal, 1975). Its unique quality is

the very fast response relative to dosing, occurring within few hours (Zarate et al., 2006;

Machado-Vieira et al., 2009). Also, the duration of ketamine’s antidepressant effects is of 3

days or more (Zarate et al., 2006; Berman et al., 2000), even though its half-life is of about

2.5 hours (Ketalar® - Medsafe). The reasons for these effects will be partially explained in

this thesis, as the action of ketamine in the brain is not fully understood. Also, the possibility

of using this medication for depressed individuals with short life expectancy, such as terminal

cancer patients, will be discussed.! !In summary, this thesis will cover several topics:

Chapter 1: introduction and epidemiology of depression;

Chapter 2: pathophysiology and treatment of MDD;

Chapter 3: literature review and meta-analyses of effectiveness of antidepressant treatments

in patients with cancer;

! %(!

Chapter 4: ketamine overview;

Chapter 5: literature review and meta-analysis of effectiveness of ketamine in MDD and

depressive episode of bipolar disorder, both case series and randomized clinical trials;

Chapter 6: description of a protocol for treating MDD in cancer patients;

Chapter 7: description of single and repeat dosing for a patient with MDD and terminal

cancer;

Chapter 8: conclusions about the possible use for ketamine as a fast acting antidepressant for

depressive episodes in terminal cancer patients.

!!1.2 History of depression!Depressive symptoms have been described since ancient history, and writings regarding them

were even found in Ancient Egyptian papyri. Also, the Bible presented King Saul with

symptoms of depression (Davison, 2009; Huisman, 2007). In ancient Greece, supernatural

explanations were provided for these symptoms, such as evil spirits and punishment from

gods, but during the 6th century BC, these explanations started losing their impact in Greek

culture.!! The first statement that melancholia (which in ancient times was described by depressive

symptoms, among others) was caused by a dysfunction of the brain was made by Hippocrates

(460- 377 BC). He believed that the excess of “black bile” in individual’s brains was

responsible for melancholic symptoms, giving rise to the humoral theory (first postulated by

Empedocles) for melancholia (Davison, 2009; Bos, 2009). However, centuries latter, the

Christian Church stated again that melancholia was a disease of the soul. This idea brought

back supernatural experiences as a possible cause for depressive symptoms, and many women

were then at risk of being considered witches and condemned to be burnt at the stake (Daly,

2007).!!In the 20th century, more plausible explanations for the symptoms of psychiatric illnesses

were raised. Emil Kraepelin adopted a somatic approach, while Sigmund Freud emphasized

psychological factors, although biological factors were also considered in his hypotheses. In

1930’s, the initiation of treatments, such as electroconvulsive therapy and psychosurgery,

raised the need for clinical categories for treatment decisions (Davison, 2009).!!!

! %)!

!1.3 Epidemiology of depression in Major Depressive Disorder and Bipolar Disorder!Patients with Major Depression and Bipolar Depression have depressive symptoms (such as

depressed mood, diminished interest in activities, diminished ability to concentrate, alteration

in appetite, fatigue, feelings of guilt, suicidal thoughts) alternately with euthymia (normal

mood). In patients with BD, episodes of hypomania/mania (usually with grandiosity, reduced

need to sleep, running thoughts, pressure to keep talking, excessive involvement in

pleasurable activities that may be harmful, distractibility and psychomotor agitation) are also

present, at some time in their lives (DSM IV).

!The yearly prevalence of MDD in general population in the United States is around 6.7%. In

New Zealand, according to The New Zealand Mental Health Survey 2003/4, almost the same

prevalence is found, with major depressive episodes (MDE) affecting 6.6% of general

population (Kessler et al., 2005; Scott et al., 2010). In the New Zealand survey, these episodes

were more commonly found in females (8.1% versus 4.9% in males). Fewer than 10% of

MDE episodes were considered mild, with over half being severe or very severe. Also, just

one-third of patients with severe impairment received treatment from mental health services.

This information may help doctors with the awareness of considerably high prevalence of

depression among patients in general population and its severity. !!Besides differences between MDD and BD, in BD depression is the most experienced mood

(Mitchell and Malhi, 2004). In both cases, there is a correlation with cardiovascular disease,

obesity, metabolic syndrome, dementia, low income, marital status, and others (Weiner et al.,

2011; Ko et al., 2010; Keddie, 2011; Fiedorowicz et al., 2008; Byers and Yaffe, 2011;

Kessing and Andersen, 2004; Schofield et al., 2011; Schoeyen et al., 2011; Yan et al., 2011).

Some associations seem to be explainable, such as lack of exercise and eating habits

associated with metabolic syndrome. However, it is hard to analyse these correlations as cause

of depression, its consequence, or as part of a broader syndrome (Kahl et al., 2011). Also,

these individuals have a higher risk of suicide, and there are more deaths from unnatural

causes in the unipolar depression group, when compared to bipolar depression (Osby et al.,

2001; Black et al., 1987). The costs of depression are high, for example, the economic burden

in USA was of almost $44 billions in 1990 (Greenberg et al., 1993). General medical care

costs are much higher than those with general population, even for depressed subjects that are

on treatment (Bosmans et al., 2010). Although this population maintains an intense use of

professional care, they have higher chances of not meeting their needs, and of having a

! *+!

chronic illness, that may be due to inadequate treatment (Bijil et al., 2000; Pincus and Pettit,

2001). Not only mental health professionals should be aware of these facts, but also general

practioners, as according to Bijil et al., 2000 and Lepine et al., 1997, the majority of

individuals with depression look for treatment in primary care. Thus the treatment for

depressive symptoms is of great importance in both mood disorders.

For the estimation of the global burden in patients with MDD (World Health Organization –

WHO), symptoms severity were split in disability weights: 0.14, 0.35, and 0.76 for mild,

moderate and severe, respectively (Ustün et al., 2004). Actually, many individuals suffer from

chronic or recurrent depression, with some of these patients achieving recovery only 6 months

after initial treatment (50% of recovery, according to Pincus and Pettit, 2001). In Major

Depression, there is less chance of recovery over time with symptomatology, and with each

subsequent episode (Lavori et al., 1994; Keller et al., 1992), leading to a high prevalence of

chronically affected patients, who make up 26.8% of patients with MDD (Satyanarayana et

al., 2009). Moreover, according to the WHO, there is an estimative that, in 2030, unipolar

depression will be the second leading cause of disability adjusted for life years. This is similar

to a previous projection made for 2020 sixteen years ago (Mathers and Loncar, 2005; Murray

and Lopez, 1996). The World Health Organization also states that MDD already comprises

10.7% of total global years lived with disability, and BD accounts for other 2.5% (Ustun et

al., 2004; Ayuso-Mateos, 2006).

Antidepressant treatment for MDD is known to be effective, even though remission rates are

considered not optimal (Khawam et al., 2006; Trivedi et al., 2006). However, its use for

depressive episodes in patients with BD is not clearly beneficial, with some authors reporting

response to antidepressants in this group (Pacchiarotti et al., 2011; Ghaemi et al., 2008) and a

recent review (Sidor and Macqueen, 2011) suggesting that antidepressants are of little help for

acute treatment of depressive episode in BD. Also, Sidor and Macqueen, 2011 state that the

risk of mania switch depends on the criteria used to evaluate it. This risk has been

continuously investigated, and includes cycle acceleration, mania, hypomania and mixed

switch (Amsterdan and Shults, 2010; Pacchiarotti et al., 2009; Ghaemi et al., 2008; Goodnick,

2007). Considering the low response in depressive episodes achieved with antidepressant

medications available, treatment alternatives are desirable.

! *%!

1.4 Epidemiology of Major Depressive Disorder in cancer patients

Depression rates in individuals with advanced cancer are very high, affecting approximately

15% of patients (Mitchell et al., 2011), and when considering subsyndromal depression (the

presence of depressive symptoms that do not meet criteria for MDD) the numbers are even

higher (Boyes et al., 2011). Although diagnoses of adjustment disorder and of minor

depression are quite common in cancer patients (Mitchell et al., 2011), initial support and

follow up of depressive symptoms is very important to ensure appropriate treatment.

Psychological support must be considered for cancer patients during their treatment. For

better results, it should start from the time of cancer diagnosis and, in addition, support for

relatives and caregivers should also be provided (McLean et al., 2011; Kang et al., 2012). !

It is important to be aware that recent data demonstrates even higher rates of depression in

patients prior to commencing chemotherapy, with prevalence as high as 25% (Breen et al.,

2009). Furthermore, suicidal ideation has been found in 10% of cancer patients, who were

more frequently females, with worse interpersonal relationships, significant pain, MDD,

dysthymia and panic disorder (Madeira et al., 2011). Therefore, a complete assessment of

previous and actual history of psychiatric illness, together with detailed interpersonal

relationships, is vital to achieving appropriate treatment (Boyes et al., 2011). Very little

information about treatment of depression was found concerning cancer patients, which will

be discussed in detail further, in Chapter 3. This fact raised concerns about the lack of

information concerning the possibility of different responses to treatment of MDD in this

specific population. !!Depressive episodes may initially start with symptoms that meet criteria for adjustment

disorder (a maladaptive reaction to a stressful life event) and early intervention is essential for

a better prognosis (Akechi et al., 2004). This situation is complicated, because many

symptoms due to cancer or its treatment are also part of MDD criteria (for example, weight

loss, fatigue, psychomotor retardation and diminished ability to concentrate). Trying to solve

this problem, many researchers have assessed mood symptoms with the Hospital Anxiety and

Depression Scale (HADS), which is extensively used in palliative care, due to its absence of

physical symptoms on its assessment. HADS is a validated and reliable tool for medically ill

patients’ assessments for anxiety and depression (Guidi et al., 2011; Castelli et al., 2011).

Also, The Demoralization Scale may be a useful instrument, offering an evaluation of non-

specific dysphoria, social disconnectedness, hopelessness, helplessness and suicidal ideation

(Kissane et al., 2004).

! **!

The impact that MDD has on these patients may result in choices that might be different if

they were not affected by the disorder. For example, a study with cancer patients showed that

acceptance of adjuvant chemotherapy in patients with breast cancer and depression was

51.3%, but among patients without MDD, 92.2% accepted the abovementioned treatment

(Colleoni et al., 2000). With this knowledge, there are more chances of considering a possible

diagnosis of depression in this population. . This awareness could also lead patients to MDD

treatment, and probably to acceptance of cancer treatments, increasing their chances of

survival or recovery.!

Considering the high prevalence of depression in this population, studies evaluating

antidepressant treatments are of great importance for the clinical practice. Unfortunately there

are only two randomized clinical trials on this topic (Costa et al., 1985; van Heeringen and

Zivkov, 1996). A number of other studies have been published, however these have combined

patients with subsyndromal depression and MDD, usually neglecting illness severity. For

patients with terminal cancer, there is an even greater lack of studies, directing attention to the

fact that more could be done for this particular group. There are many studies considering

subjects support via telephone and via internet (White et al., 2011; Klemm and Hardie, 2002),

but usually these trials are for cancer survivors and no similar study was found for advanced

cancer patients.

! *,!

CHAPTER 2

Pathophysiology and treatment of Major

Depressive Disorder

! *-!

2.1 Pathophysiology of Major Depressive Disorder

Depression has been recognized since ancient times but is still considered a complex disease

with unknown etiology (Dagyt! et al., 2010). For many years the monoaminergic system was

the focus for depression treatment. However, with available antidepressants (which target

monoamines), patients’ response is very limited. This fact led researchers to consider the

possibility that other alterations in depressed subjects’ brains might be involved in the

pathophysiology of MDD. In the last decades, hippocampal neurogenesis started to be

investigated in depression’s pathophysiology (Dagyt! et al., 2011), and many hypotheses have

been studied as an attempt to better understand it.

The hypotheses described below are interconnected, indicating that depression is a highly

complex illness. For example, the locus coeruleus, which is considered a part of the central

stress system, has noradrenergic innervation, and is usually overexcited and may be defective

in patients with MDD. (Itoi and Sugimito, 2010; Harro and Oreland, 2001). When the locus

coeruleus is activated, it fires the prefrontal cortex (PFC), and triggers the ventral tegmental

area (VTA), releasing dopamine in the nucleus accumbens (NA). The PFC relays to the VTA,

releasing glutamate in the last, leading to increased excitability and more dopamine release in

the NA (Sara, 2009). This illustrates the interaction between different systems. Further,

cytokines (through influences on synaptic plasticity, and on neuroendorine and monoamine

systems) can influence corticotropin-releasing hormone (CRH), brain-derived neurotrophic

factor (BDNF), serotonin, norepinephrine, and dopamine (Miller and Raison, 2008). Figure

2.1 illustrates these interactions. The following sections will discuss some of the hypotheses

that try to explain the neurobiology of depression.

! *.!

Figure 2.1: Anatomical connections and interactions between neuroendocrine system, BDNF,

monoamines, and cytokines in the brain.

Figure modified from: www.antisocial-disorder.com/page/9

VTA

NA

PFC

!!!!!!!! LC

! *&!

2.1.1 Serotonergic system

The monoamine hypothesis of depression, which proposed that decreased levels of serotonin,

noradrenaline and/or dopamine would induce depressive symptoms (Middlemiss et al., 2002),

was previously converged to the serotoninergic system as a possible main alteration in the

brain, leading to depression (Coppen, 1967). Also, serotonin’s precursor tryptophan, proved

to be decreased in subjects with MDD (Anderson et al., 1990). Treatment with tryptophan had

a positive effect on mood when administered to depressed patients (Young et al., 1985;

Shopsin, 1978), suggesting a role for serotonin in depressive symptoms. Also, an immune-

response with consequent depletion of tryptophan has been associated with depression in

cancer patients (Kurz et al., 2011). Another fact that contributes to the association between the

serotonergic system and depression is the fact that many patients with MDD present chronic

pain symptoms. Regarding this fact, as the serotonergic 5-HT1A receptor seems to play a role

in Major Depression and in pain regulation (Ohayon, 2009), serotonin could provide a link

between these symptoms, and explain their frequent coexistence.

2.1.2 Noradrenergic system

This system is considered to be related to several areas that are altered in subjects with MDD,

such as attention, memory, learning, sleep, emotion and response to stress (Sara, 2009;

Berridge and Waterhouse, 2003). It is also implicated in synaptic plasticity in the

hippocampus (Sara, 2009), possibly playing a role in MDD. Its dysfunction has constantly

been pointed as a possible mechanism in depressive symptoms (Goekoop et al., 2011; Itoi and

Sugimoto, 2010). Some antidepressants with action in the noradrenergic system have been

cited as possibly more effective treatments when compared to selective serotonin-reuptake

inhibitors (SSRIs) however, this is still controversial. Besides these medications also act in

the dopaminergic system, some authors correlate their noradrenergic action to a possible

pathway to the treatment of Major Depression (Montgomery and Briley, 2011; Nutt et al.,

2007).

The noradrenergic system is linked to the activation of the hypothalamic-pituitary-adrenal

(HPA) axis, and vasopressin release, which are also implicated in the pathophysiology of

depression (Keller et al., 2006; Goekoop et al., 2011).

! *'!

2.1.3 Dopaminergic system

The role of dopamine in Major Depression seems to be highly linked to attention,

psychomotor activity, and anhedonia (which is the inability of experiencing pleasure in

activities that before were enjoyable). Because many antidepressants do not directly act in the

dopaminergic system, this might contribute to the residual symptoms that many patients

experience during their treatment. Residual symptoms frequently consist of impaired

concentration, motivation and pleasure (Dunlop and Nemeroff, 2007; Nutt, 2006). In the

cerebrospinal fluid of depressed subjects, homovanillic acid (HVA) seems to be decreased.

Some antidepressants are likely to act in the dopaminergic system, such as sertraline,

bupropion and the monoamine oxidase inhibitors (MAOIs), but the magnitude of this action

still requires further studies (Boadie et al., 2007).

2.1.4 HPA axis

The hyperactivity of the hypothalamic- pituitary-adrenal (HPA) axis is the main stress system

in the human body, and is responsible for stress response adaptation. It is related not only to

hormones and peptides, but also to neurotransmitters, such as noradrenaline, dopamine and

serotonin (Joëls and Baram, 2009). The hyperactivity of the HPA axis, which is illustrated in

figure 2.2, induces an exacerbated release of corticotropin-releasing factor (CRF), thus

secreting more ACTH and leading to hypercortisolemia (Keller et al., 2006). Also, HPA axis’

activation is considered to be involved in stress-induced neuronal atrophy. Two types of

neuronal plasticity are impacted by chronic stress, with atrophy of dentrites in the CA3 region

and repressed neurogenesis of granule cells in the dentate gyrus, in which not only

glucocorticoids but also NMDA receptors play a role (McEwen, 1999). Thus, chronic

psychosocial stress may lead to inhibition of neurogenesis and even neuronal loss (McEwen

and Mangarinos, 2001), which may contribute to cognitive impairment in MDD.

! *(!

Figure 2.2: HPA axis dysfunction in MDD.

Hypotheses for HPA axis dysregulation:

(a): Glucocorticoid receptor (GR) hypothesis: when patients with MDD have to deal with stressful events in their

lives, the GR resistance and the reduced negative feedback in depression are the predominant causes of the

elevation of corticotropin-releasing factor (CRF), adrenocorticotropine (ACTH) and cortisol.

(b): CRF hyperdrive hypothesis (consists on multiple feedback loops):

loop 1: the excess of cortisol leads to a downregulation of GR, which fails to contain the hyperactivity of the

HPA axis.

loop 2: CRF may able to enhance its own biosynthesis in the paraventricular nucleus (PVN) of the

hypothalamus.

loop 3: continuous activation of the HPA axis up-regulates the amygdaloid CRF system, stimulating the HPA

axis in turn.

Figure copied from: van Den Eede and Claes, 2004.

! *)!

2.1.5 Gabaergic system

Gamma-Aminobutyric acid (GABA) is the most common inhibitory neurotransmitter in the

human brain, and glutamate is involved in GABA’s synthesis. This system is involved in

mood disorders, and increased GABAergic tone may lead to antidepressant effects (Brambilla

et al., 2003). In the cerebrospinal fluid, and in the plasma, depressed patients have lower

levels of GABA (Petty et al., 1992; Berrettini et al., 1983). However, some studies found that,

even after antidepressant treatment, plasma levels of GABA remain low. Another finding was

that level alterations were not associated with depression severity (Petty et al., 1992, 1993).

Different studies suggest that lower GABAergic functioning may be involved in the

pathophysiology of depression (Holland et al., 2010; Zink et al., 2009). With all positive

evidence, the focus on GABAergic system has also contributed to research for new

antidepressant treatments (Mohler, 2012).

2.1.6 Glutamatergic system

Glutamate is the most abundant excitatory neurotransmitter found in the brain, acting in the

“tripartite glutamatergic synapse” (Figure 2.3), involving different glutamate receptors and

other targets with therapeutic potentials (Popoli et al., 2011; Zarate et al. 2010). A number of

studies have used Magnetic Resonance Spectroscopy (MRS) as an attempt to assess activity

of the glutamatergic system in specific regions in the brain (Sanacora et al., 2004; Hasler et

al., 2007; Zarate et al., 2010). These studies have reported that these measures vary

considerably by brain region, type of mood disorder, course, progression and phase of illness

(Sanacora et al., 2012). Glutamate receptors are classified as metabotropic and ionotropic, the

last one includes N-methyl-D-aspartate (NMDA), which is the receptor linked to ketamine

action as a fast acting antidepressant (Paul and Skolnick, 2003).!!!!!!!!!!

! ,+!

Figure 2.3: Tripartide glutamatergic synapse.!!!

! Neuronal glutamate (Glu) is synthesized from glucose and glutamine (Gln), which is provided by glial cells. Then, vesicular glutamate transporters (vGluTs ) pack glutamate and SNARE complex proteins mediate the merge of vesicles in the presynaptic membrane. In the extracellular space, glutamate is tied to:

• ionotropic glutamate receptors (NMDA receptors - NMDARs) • AMPA receptors (AMPARs) • metabotropic glutamate receptors (mGluR1 to mGluR8)

While connected, the receptors initiate various responses, and glutamate is cleared from the synapse through excitatory amino acid transporters (EAATs) on neighbouring glial cells (EAAT1 and EAAT2) and on neurons, in lower intensity (EAAT3 and EAAT4). Within the glial cell, glutamate is converted to glutamine by glutamine synthetase, restarting the glutamate–glutamine cycle. Figure from: Popoli et al., 2012.

! ,%!

2.1.7 Cytokines

Cytokines are signalling molecules produced by immune cells and are involved in regulating

immune responses. Currently, these molecules have been frequently associated with

depression and have been possibly linked to anorexia, found in several depressed patients

(Schiepers et al., 2005; Anderson, 1996). The communication between the central nervous

system and the immune system has lead to a research focus on cytokines’ relationship with

MDD. Also, several illnesses that present chronic inflammatory response are frequently

known to coexist with MDD. Further, it is well established that there is an association

between depressive symptoms and the use of pro-inflammatory cytokines, which are

frequently used for the treatment of cancer (Yirmiya at al., 1999; O’Connor et al., 2007).

Several factors are related to the role of inflammation in the pathophysiology in depression

(Leonard and Maes, 2012), as demonstrated in Figure 2.4.

Figure 2.4: From inflammation to depressive symptoms.

Peripheral cell-mediated immune activation leads to increased interferon (IFN), interleukin-1 (IL-1), interleukin-2 (IL-2), interleukin-6 (IL) and tumor necrosis factor (TNF). With indoleamine (IDO) activation , a depletion of l-tryptophan occurs, leading to depressive symptoms. Also, the production of tryptophan catabolites (TRYCATs) induces depression.. At the same time, there is the iduction of upregulation of the 5-HT transporter (SERT), diminishing synaptic dopamine (5-HT), which also induces depressive symptoms. !Figure from: Leonard and Maes, 2012.!

!

! ,*!

2.2 Current treatments for depressive episodes

This topic briefly covers the main available treatments for MDD, including some options for

treatment resistant depression.

2.2.1 Psychotherapy

It is widely accepted that psychotherapy is a very important tool in the treatment of several

psychiatric disorders. The most studied are interpersonal, psychodynamic, cognitive and

behavior therapy (Markowitz and Weissman, 2012; Feng et al., 2012; Abbass and Driessen,

2010). Differences found in responses to psychotherapeutic treatments are minor, and overall

these approaches appear to be as effective as antidepressant medications for mild and

moderate depressive episodes (Cuijpers et al., 2011; Härter et al., 2010).

Psychotherapy should be recommended for acute treatment of depression, together with

antidepressant pharmacotherapy, as it may lead to improvement in patients’ quality of life. In

treatment resistant depression results are more varied, nevertheless psychotherapy (cognitive,

interpersonal and behavioral) should be indicated for these individuals (Ishak et al., 2011;

Jakobsen, 2011; Trivedi et al., 2011).

This treatment together with pharmacotherapy usually takes a few weeks to result in

significant response (van Calker et al., 2009). Although psychotherapy is very important for

patients with cancer (Savard et al., 2006), individuals with end-stage cancer may not fully

benefit from this treatment, as their life expectancy is very short.

2.2.2 Pharmacotherapy

Antidepressants are the main treatment for moderate and severe MDD. The selective

serotonin reuptake inhibitors (SSRIs), due to their favorable safety and efficacy profiles, are

usually the first line antidepressants to be indicated for depressive episodes. Also, although

there are several SSRIs, minimal differences can be found amongst them (Souery et al., 2011;

Boulenger et al., 2010). According to some authors, serotonin-norepinephrine reuptake

inhibitors (SNRIs) are broadly similar to SSRIs in terms of efficacy and safety. Their rates of

response and remission are similar, (Weilburg, 2004; Kornstein et al., 2009). Older agents,

such as tricyclic antidepressants and monoamine oxidase inhibitors, despite being effective,

present more side effects. Even, monoamine oxidase inhibitors (MAOIs) present high

! ,,!

toxicity, and these agents, together with tricyclic antidepressants, have higher incidence of

withdrawal when compared to SSRI (Chouinard et al., 1994; Amsterdam and Shults, 2005).

Table 2.1: Main mechanisms of action of antidepressants.

MONOAMINE REUPTAKE INHIBITORS

Serotonin selective SSRIs, Clomipramine

Norepinephrine selective Reboxetine, Maprotiline, Desipramine

Dopamine selective Bupropion

Mixed reuptake inhibitors Venlafaxine, Imipramine, other tryciclics

RECEPTOR ANTAGONISTS

Mianserin, Mirtazapine (5HT2A, 5HT2C, alpha-2)

ENZYME INHIBITORS

Irreversible MAOIs Phenelzine, Tranylcypromine

Reversible MAOIs Moclobemide

2.2.3 Electroconvulsive therapy (ECT)

ECT is a validated treatment for several psychiatric disorders, and consists of

neurostimulation by the induction of a grand mal seizure. ECT’s effects seem to rely in

changes in the monoaminergic system and in the stress response system. It was first described

in 1930’s and over time its efficacy and safety have been extensively studied. ECT’s efficacy

is equally consistent for unipolar and for bipolar depressive episodes. (Trevino et al., 2010;

Nikisch and Mathé, 2008; Coentre et al., 2009; Bailine et al., 2010). It is usually administered

from 1 to 3 times a week, and a meta-analysis with 73 randomized clinical trials showed that

there is no difference in efficacy between these time-points (UK ECT Review group, 2003).

Although cognitive impairment is more significant for bilateral and high dose ECT, when

compared to unilateral and brief pulse, it is also more effective than lower dose treatment.

Also, for continuation electroconvulsive therapy, time between session is increased, which

reduces the chances of cognitive side effects (Lisanby et al., 2000; UK ECT Review group,

2003; Trevino et al, 2010).

! ,-!

2.2.4 Repetitive transcranial magnetic stimulation

Transcranial magnetic stimulation (TMS) was first described in 1985, and consists in an

intense and brief electromagnetic field. It is a noninvasive procedure that is easily tolerated.

(Dell'osso et al., 2011; Gershon et al., 2003).

Repetitive TMS is the most used modality of TMS in psychiatry, providing more prolonged

results than the single-pulse modality. Also, as ECT, it acts on neuroendocrine and

neurotransmitters systems (Baeken and De Raedt, 2011). Treatments are made in a daily basis

for 5 days/week, usually for about 2 to 9 weeks. (Dell'osso et al., 2011; Fitzgerald and

Daskalakis, 2011). Side effects are less significant than those found in ECT, such as memory

impairment, which is usually not present in subjects who received repetitive TMS, except for

those who presented a seizure during treatment, which is not a common side effect

(Wassermann, 2000; Loo et al., 2008).

Comparatively, although repetitive TMS has fewer side effects than ECT, the latter has higher

rates of remission. This fact emphasizes the utility of ECT for treatment resistant patients

(Minichino et al., 2012).

2.2.5 Vagus nerve stimulation (VNS)

VNS was originally developed as a treatment for patients with epilepsy that did not respond to

pharmacological treatment. Recent data has implicated it as a possible treatment for

individuals with depression, although these studies are not controlled. Also, there is the

possibility that it plays a role in serotoninergic and noradrenergic systems as a mechanism for

antidepressant response (Martin and Martín-Sánchez, 2011; Dorr and Debonnel, 2006). A

recent meta-analysis showed that, although several studies demonstrate that VNS may be used

as a possible treatment for depression, its efficacy is still questionable and it may actually

represent a placebo effect (Martin and Martín-Sánchez, 2011). However, a positive aspect of

VNS is the lack of cognitive side effects, specially when compared to ECT. As positive mood

effects were related to subjects who did not have a high degree of resistance to previous

treatments, the use of VNS requires careful consideration (Sackelm et al., 2001).

! ,.!

2.2.6 Deep brain stimulation (DBS)

DBS is an invasive procedure, which involves the insertion of stimulating electrodes in the

brain. Its use is still experimental for depression, although its use for Parkinson’s disease and

refractory obsessive compulsive disorder is approved by the FDA. It consists of a 2 phase

procedure, with the first one performed under local anesthesia and with the patient awake, as

the effects od stimulation must be evaluated. In the second phase, the inserction of a pulse

generator in the subcutaneus fat, under the patient’s subclavian region, is made under general

anesthesia. Then it is connected to cables under the skin, that are connected to the electrodes

in the brain (Bell et al., 2009; Malone, 2010; Blomstedt et al., 2010). Studies show positive

outcomes for patients with treatment resistant depression, however its action in the brain is

not completely clear (Hamani and Nobrega, 2010). In 2005, Mayberg et al. used DBS for

patients with treatment-resistant depression, achieving a sudden and sustained improvement

in mood, in 66% of these patients. The change in mood seems to be related to a fast

deactivation of Broadmann area 25, which is hyperactive in MDD (Mayberg et al., 2005;

1999). However, there are various risks involving this procedure, such as stroke, infection,

paresthesia, and anxiety (Holtzheimer and Mayberg, 2010; Malone, 2010; Blomstedt et al.,

2010). Thus, severity of refractory depression should be carefully evaluated before

considering this treatment.

! ,&!

CHAPTER 3

Review and meta-analysis of available

treatments for depression in cancer patients

! ,'!

3.1 Introduction

As stated in chapter one of this thesis, Major Depressive Disorder (MDD) has a high 12-

month prevalence in general population, and higher rates are seen in patients with advanced

cancer (Scott et al., 2010; Kessler et al., 2005). In addition, depression is the main mental

health complication of cancer (Mitchell et al., 2011).

In patients with cancer, those with past history of Major Depression have a higher probability

of developing depressive episodes after a diagnosis of cancer. As this finding implicates the

possibility of early diagnosis and intervention, depression should be considered a special topic

in psycho-oncology and not be misdiagnosed (Mitchell 2011; Mitchell et al., 2011). More

over, this specific population appears to have a lower level of acceptance of adjuvant

chemotherapy (Colleoni et al., 2000), and in those who will commence chemotherapy,

prevalence rates of MDD may be as high as 25% (Breen et al., 2009). For patients with

cancer, depression can be not only a burden, but also a risk, when considering their

psychological symptoms may affect decisions for accepting treatment options.

Proper attention and treatment should be given to these individuals, as antidepressant drugs

are effective in palliative care settings. However, these medications seem to be

underprescribed for older patients and for those with physical illnesses, despite having MDD

(Rayner et al., 2011). The objective of this chapter is to investigate the safety and

effectiveness of currently available antidepressant treatments in cancer patients.

3.2 Methods

3.2.1 Search strategy

Electronic databases used were: ClinicalTrials.gov, PsychInfo and Pubmed. In all three

databases, search was made with the keywords “treatment” [AND] “depression” [AND]

“cancer”. In Pubmed, the limit “Randomized Controlled Trial” was applied. Articles from

1971 until January 2012 and in English, Portuguese, Spanish and Italian were reviewed. Also,

citations from screened articles were verified.

3.2.2 Selection criteria

For inclusion in the review, studies had to meet the following criteria:

• double-blind Randomized Controlled Trial (RCT),

! ,(!

• placebo controlled,

• presence of at least 20 subjects in each arm,

• patients fulfilling DSM (Diagnostic and Statistical Manual of Mental Disorders) or

ICD (International Classification of Disease) criteria for depressive episode,

• patients with cancer,

• absence of initial antidepressant treatment (or change in current antidepressant

treatment) at the same time of intervention,

• psychopharmacologic or psychotherapeutic treatment for depression.

Any type of psychotherapy was considered in this search.

Figure 3.1: Selection of trials for meta-analysis.

!!

20 articles were screened

18 articles excluded (did not meet criteria): 12 not all patients with MDD 4 individualized treatment (non-randomized) 1 no placebo group 1 psychotherapy trial did not exclude use of antidepressant medication

2 pharmacological trials

included in review

0 psychotherapy trials included in

review

Search of electronic databases: ClinicalTrials.gov (346 articles) PsychInfo (34 articles) Pubmed (633 articles) !

! ,)!

3.2.3 Analysis

The selected trials for meta-analysis were analyzed with RevMan 5.1 (Cochrane

Collaboration, 2008). Data for the outcomes for treatment versus placebo were statistically

examined as continuous data, with random effects model, and standardized mean difference

for effect measure. The I2 statistic was used to assess heterogeneity of studies.

3.3 Results

After a systematic search 20 articles were selected for review, of which only 2 met criteria for

meta-analysis (Costa et al., 1985; van Heeringen and Zivkov, 1996). Both trials investigated

the use of pharmacotherapy for depression in cancer patients, and no psychotherapy trial met

criteria for meta-analysis. Among the screened studies, several did not meet criteria for MDD,

although these quoted “depression”, instead of “depressive symptoms” or “adjustment

disorder” in some cases (Bruera et al., 1986; Holland et al., 1998; Roscoe et al., 2005; Savard

et al., 2006; Bar-Sela et al., 2007). In other trials, patients were offered individualized

treatments, thus not appropriate for comparison with other therapies (Ell et al., 2011; Ell et al.,

2008; Strong et al., 2008; Dwight-Johnson et al., 2005). One study compared fluoxetine and

amitriptyline for cancer patients, although it met criteria for MDD according to ICD-10 it was

not included in meta-analysis because of its lack of placebo group (Pezzella et al., 2001). A

psychotherapy trial was excluded because it was not clear if patients could use antidepressant

medications or not (Evans and Connis, 1995).

3.3.1 Meta-analysis

Both of the selected articles compared mianserin (a tetracyclic antidepressant) to placebo,

both evaluated its response in female patients (van Heeringen and Zivkov, 1996; Costa et al.,

1985). Costa et al., 1985 included subjects with general gynecological cancer, and the second

study assessed patients with breast cancer. Another difference between studied populations

was the cancer’s severity, with Costa et al., 1985 including more severely ill patients (several

of them with terminal cancer).

While Costa et al., 1985 showed positive results within 7 days after the initiation of the use of

mianserin, van Heeringen and Zivkov, 1996 reported positive outcome starting on day 14

post-treatment. In the first week patients received 30 mg/day of mianserin, in the following

weeks the dose was increased to 60 mg/day. Mianserin was given to patients three times a day

! -+!

in the trial from Costa et al., 1985 and once a day in van Heeringen and Zivkov, 1996. In

these trials, there were no significant differences in side effects when comparing intervention

and control group. Also, higher rates of premature terminations were observed in controls,

and these were linked to lack of efficacy. Mianserin 60 mg/day was safe when used together

with chemotherapy and with radiotherapy in both trials. The low I2 value (0%) suggests lack

of heterogeneity in these two sets of clinical trial data. The forest plot of these two studies is

shown in figure 3.2, and indicates a moderate statistically significant effect size (SMD= -

0.68), p= 0.0002.

Figure 3.2: Meta-analysis of antidepressant trials in patients with MDD and cancer (HAMD).

3.4 Discussion

Although there are many studies considering depressive symptoms and their treatment in

cancer patients, most of these did not include patients with Major Depression. These studies

frequently included patients with mild symptomatology and/or adjustment disorder (Bruera et

al., 1986; Holland et al., 1998; Roscoe et al., 2005; Savard et al., 2006; Bar-Sela et al., 2007),

making it impossible to analyse treatment response for MDD in this population. After

applying selection criteria, only two studies were suitable for analysis (van Heeringen and

Zivkov, 1996; Costa et al., 1985). This shows a deficit of appropriate research in this area,

which was an unexpected result. Other reviews that presented similar objectives to this

chapter were then assessed, and the included trials in these studies did not meet criteria for

Major Depression (Akechi et al., 2008; Williams and Dale, 2006). All the studies that were

excluded because of individualized treatment addressed patients with MDD, usually with mild

depression. In these trials, treatments included psycho-education, psychotherapy, and the use

of antidepressants (Ell et al., 2011; Ell et al., 2008; Strong et al., 2008; Dwight-Johnson et al.,

2005). Also, the type of treatment offered to the patient varied according to symptomatology,

patient’s choice and level of social support. As provided treatments were different for each

! -%!

individual, it was not possible to make an analysis comparing the results from these studies

with the data available from other research.

The time of onset of MDD plays an important role in the treatment of depression, and chronic

illness and lack of response to previous treatments may occur, resulting in individuals with

more severe symptomatology and lower response to treatment (Rush et al., 2006). In the

selected studies for meta-analysis, Costa et al. analysed subjects with Major Depression

succeeding or paralleling cancer diagnosis, while van Heeringen and Zivkov, 1996 did not

consider the time of onset of depression. Thus, the second trial may have included patients

with chronic or potentially treatment resistant depression as exclusion criteria were not

described. Nevertheless, the statistical analysis shows no heterogeneity in the outcomes of the

compared studies. Both used Hamilton Depression Rating Scale (HAMD), but the numbers of

items used was different in each trial, with 17 items in Costa et al., 1985, and 21 in van

Heeringen and Zivkov, 1996. To manage this, standardized mean differences were used as the

effect measure.

Mianserin was significantly more effective than placebo in both studies, and adverse events

were similar to control group, despite use of cytotoxic drugs. These findings support the use

of mianserin in depressed patients with cancer, even for those who are receiving aggressive

treatment for malignant neoplastic disease. The fact that Costa et al., 1985 prescribed the

medication 3 times a day may reduce patient’s adherence when they are outpatients (Santiago-

Rodriguez et al., 2002). Thus, van Heeringen and Zivkov, 1996 presented a better plan of

administration, with once-a-day dosing, as mianserin’s half-life is 21 to 61 hours and justifies

this posology (Tolvon®-Medsafe). Besides the fact that Costa et al., 1985 reported

statistically significant improvement in the intervention group after 7 days, when evaluating

response of treatment as a reduction in 50% from baseline scores, both trials present response

to mianserin in day 14 after treatment initiation. Neither of the studies presented remission

(HAMD 21 and 17 " 7) of symptoms during treatment (Rudolph and Feiger, 2000;

Zimmerman et al., 2004).

3.5 Conclusions

Depression is a disorder that is common amongst patients with terminal cancer, however there

are only two treatment trials in this population. Mianserin seems to be a safe option for this

! -*!

group, however its efficacy for remission of depressive symptoms is still not clear.

Considerable additional research in this population is required.

! -,!

CHAPTER 4

Ketamine overview

! --!

4.1 Ketamine

Ketamine is an NMDA antagonist with a range of medical and non-medical uses. These will

be reviewed in the following section. These different use patterns are associated with different

doses (see Figure 4.1).

Figure 4.1: Relation between ketamine and its indications.

Figure from: Glue et al., 2011.

4.2 Ketamine as an anesthetic

Ketamine is a general anesthetic that produces dissociative anaesthesia and is generally used

in children, with doses that vary from 9- 13 mg/kg when given via intramuscular injection

(Ketalar® - Medsafe). It has been used since 1970, most extensively during wars, as it does

not require mechanical ventilation when few resources are available (Bonanno, 2002).

4.3 Ketamine as a drug of abuse

Due to its dissociative effects, some people consume it as a drug of abuse, known as “K”,

“Special K” and “Cat Valium”, but it has not been associated with dependence syndrome

(Britt and McCance-Katz, 2005). There are two extremes related to experiences reported by

subjects with a history of ketamine abuse: when the substance is used in lower doses, there is

the "K Land", a feeling of extreme relaxation; while at high doses they may experience the

"K-hole", which is like a near-death experience, and the feeling of being at high speed in a

tunnel, usually described as not a pleasurable experience (Britt and McCance-Katz, 2005;

Dillon et al., 2003). In addition, ketamine abusers frequently report: lack of coordination,

blurred vision, confusion, visual hallucinations, and dizziness.!

CORRESPONDENCE

Dose- and Exposure-Response to Ketamine inDepression

To the Editor:

W e read with interest the recent report on the repeated-dose use of ketamine as a rapidly acting antidepressant inpatients with treatment-resistant depression (1). Despite

the promising antidepressant responses to this intervention in thisand other studies (2– 4), several important technical questionsabout administration of ketamine have not yet been explored (5).These include route of administration, dose– exposure relationship,and exposure– and dose–response information. In this letter, wepresent a simulation on projected exposures between intramuscu-lar and intravenous (IV) ketamine to establish comparable dosesand report preliminary dose–response data in patients with treat-ment-refractory bipolar II depression.

Route of Administration and Dose–Exposure Rela-tionship. Most published reports on ketamine used as an anti-depressant are at a dose of .5 mg/kg, given as a slow IV infusion over40 min. This regimen is described as being based on pilot studies inhealthy volunteers (6). An intramuscular (IM) route of administra-tion has the advantage of not requiring an IV infusion pump andfacilitates dosing in patients with poor venous access.

We developed a model for IM and IV pharmacokinetic profilesbased on published reports for racemic ketamine (7,8). The IV pa-rameter values are as reported in Ihmsen et al. (8), and relativebioavailability of IM ketamine to IV was 93% (7). Our model de-scribed similar overall exposures over time based on comparable IMand IV dosing (2-hour area under the concentration-time curvevalues ! 20.9 and 21.7 mg.min/L, respectively); however, peakexposures were higher after IM dosing (Figure 1A). Higher IM or IVketamine doses would be expected to demonstrate dose-propor-tional pharmacokinetics (8). We propose that IM ketamine may bean acceptable alternative route of administration for ketamine inpatients with refractory depression.

Exposure–Response Relationship. There is some sug-gestion that mood response may be related to the early ketamineexposure profile, given the substantial early improvement in de-pression ratings by 40 min after the start of the IV infusion (1– 4).

Therefore, we hypothesize that early exposure is critical to thetherapeutic effect of ketamine, and in this context, the higher peakvalues after IM ketamine may be beneficial (Figure 1A).

Dose–Response Relationship. We have preliminary dose–response data in two female patients with refractory depression,treated with open-label ascending doses of IM ketamine as part ofclinical care. An initial dose of .5 mg/kg produced minimal (10%–20%) reductions in Montgomery–Asberg Depression Rating Scale(MADRS) scores. Higher doses (.7 and 1.0 mg/kg IM) produced pro-portionally greater reductions in MADRS scores, in one case provid-ing scores consistent with remission (" 7; Figure 1B; data are forMADRS ratings 24 hours postinjection). All doses were safe. Re-ported side effects (principally light-headedness, sedation, disso-ciative symptoms) were qualitatively similar to those reported afterIV administration (1– 4). With ascending doses, there was no differ-ence in the time of onset or duration of symptoms; however, sub-jective assessments of symptom intensity indicated greater inten-sity with higher ketamine doses. It is important to note that thedoses used for antidepressant effects are similar to those used foranalgesia (9) and are approximately one tenth of that for anesthesiavia IM administration (10) (Figure 1C). Doses reported in the drugabuse literature fall into an intermediate range (11).

On the basis of published clinical rating data and our simulation,it is clear that early exposure to ketamine is important in its thera-peutic effects, and this can be achieved even more rapidly with IMdosing. Future studies should concentrate on the early exposure–response profile and its relationship with symptom response.

Paul Gluea*Abhishek Gulatib

Martin Le Nedelecc

Stephen Duffullb

Departments of aPsychological Medicine, bPharmacy, and cPharmacology, Universityof Otago, PO Box 913, Dunedin, New Zealand.

*Corresponding author E-mail: [email protected].

None of the authors have any biomedical or financial interests or potential conflicts ofinterest.

Please also see associated correspondence, doi 10.1016/j.biopsych.2011.02.018.

conc

entra

tion

(mg/

L)

time (mins)0 20 40 60 80 100 120

0.0

0.2

0.4

0.6

0.8IV:IM:

Ketamine dose (mg/kg IM)Baseline 0.5 0.7 1.0

MA

DR

S s

core

0

10

20

30

40 Subject 1Subject 2

A

Ketamine dose (mg/kg)0 2 4 6 8 10 12 14

Anesthesia (IM)Analgesia Abuse

DepressionC

B

Figure 1. (A) Simulation of concentration-time profilesup to 120 min for ketamine .5 mg/kg, after intramuscular(IM; dotted line) or intravenous (IV; solid line) administra-tion. (B) Effects of ascending ketamine doses on Mont-gomery-Asberg Depression Rating Scale scores in twodepressed patients, 24 hours postinjection. (C) Ketaminedose–indication relationship.

BIOL PSYCHIATRY 2011;70:e9–e100006-3223/$36.00© 2011 Society of Biological Psychiatry

!

! -.!

Some studies were conduced to assess the effects of this medication when used repeatedly as

a “club drug” on a long-term basis. However, as concomitant use of other drugs and alcohol is

very common, the results of possible cognition impairment may be due to other substances

(Dillon et al., 2003). A finding that is consistent with prolonged ketamine use, is severe lower

urinary tract symptoms, with inflammation and hematuria, many times leading to a diagnosis

of ulcerative cystitis at biopsy, which shows slow response to treatment. Although it is not a

common complication (Mason et al., 2010; Mak et al., 2011), the use of pentosane polysulfate

(Shahani et al., 2007), seems to be the best option as a treatment for ulcerative cystitis,

together with ketamine cessation. Also, ketamine is often prescribed as an analgesic, and has

been studied as a fast acting antidepressant.

4.4 Ketamine as an analgesic

Low doses of ketamine are used for analgesia, commonly between 0.1 and 2 mg/kg. Its use as

an analgesic has been studied in children, patients with chronic pain, patients in Emergency

Departments, and in postoperative settings. (Laskowski et al., 2011; Dadu et al., 2011; Lester

et al., 2010; Deng et al., 2009; Kwok et al., 2004). The duration of analgesic effects closely

follows ketamine plasma concentrations. Thus, most studies have found a significant duration

in early (30 minutes to 4 hours) pain scores, but some authors have also demonstrated pain

relief in late (24 to 72 hours) pain scores (Noppers et al., 2011; Laskowski et al., 2011; Dadu

et al., 2011; Kwok et al., 2004).

A recent meta-analysis concluded that ketamine improves the quality of pain control, and

decreases opioid consumption (Laskowski et al., 2011).

4.5 Ketamine as a fast acting antidepressant

Ketamine, a noncompetitive antagonist of the glutamate receptor NMDA (N-methyl-D-

aspartate), has been suggested as a possible option for treatment resistant Major Depression

(Berman et al., 2000; Zarate et al., 2006; aan het Rot et al., 2010).!

The mechanism of ketamine’s action on mood is not completely clear, but a signaling cascade

leading to synaptogenesis has been studied (Duman et al., 2012). Phelps et al. (2009) have

suggested a better response in patients with familial history of alcohol dependence, because

! -&!

its pathophysiology is also linked to the glutamatergic system. The correlation between

depressive symptoms and pain has not been assessed during ketamine use for depression.

In addition to the rapid onset of antidepressant action, there is a sustained response as well,

for up to one week. This appears to be unrelated to presence of ketamine (half life ~ 3 hours)

or its weakly active metabolite norketamine (half life ~ 2 hours). Recent research suggests

this may be attributable to changes in secondary signaling, and not via receptor blockade.

Therefore, the activation of a protein kinase, mammalian target of rapamycin (mTOR), may

exert the sustained antidepressant effect by the formation of new synapses – synaptogenesis.

Also, mGlu2/3 receptors, seem to activate mTOR signalling, but do not seem to be involved

in ketamine’s acute antidepressant response (Koike et al., 2011).

/01! 21345631! 75! 8179:"61! 5;;<23! within hours or days, usually with maintenance of

response for up to seven days (Zarate et al., 2006; Liebrenz et al., 2009). The doses studied to

treat Major Depressive Disorder (MDD) are low, around 0.5mg/kg, and ketamine’s

administration is usually made with intravenous infusion over 40 to 60 minutes (Machado-

Vieira et al., 2009). Nevertheless, dosage and administration can vary, usually from 0.2mg/kg

to 1.5mg/kg, and administration can also be oral and intramuscular (Goforth and Holsinger,

2007; Irwin and Iglewicz, 2010; Paslakis et al., 2010; Messer et al., 2010; Larkin and

Beautrais, 2011; Glue et al., 2011). Main side effects found with this treatment are dizziness,

psychotomimetic and dissociative effects, which are transient and moderate, dissipating in 80

minutes post infusion (Zarate et al., 2006). !

4.6 The possible use of ketamine as a fast acting antidepressant for terminal cancer

patients

Considering terminal cancer patients are in their last days or months of life, an improvement

in quality of life would mean much, not only for those who are sick, but for their families,

who also suffer greatly. Even, the fact that many people with a diagnosis of cancer present

pain related to their illness, ketamine may be effective for both, depressive symptoms and

pain, as it is frequently used for pain relief in palliative care settings.

!

! -'!

CHAPTER 5

Review and meta-analysis of ketamine as a

fast acting antidepressant

! -(!

5.1 Introduction

Major Depressive Disorder (MDD) is a common disease, with a 12-month prevalence of 6.7%

and a lifetime prevalence of 16.2% (Kessler et al., 2005). A recent study showed that only

around 33% of patients with severe impairment due to MDD receive treatment in mental

health services, indicating that the majority of patients with MDD lack proper treatment and

follow up (Scott et al., 2010). Even more, in 2004, the World Health Organization (WHO)

determined Major Depressive Disorder (MDD) as the main global cause of years lost due to

disability. This data shows that there are major, social and economic, impacts related to MDD

worldwide.

The classical understanding of MDD neurobiology focused on the monoamines, such as

serotonin, norepinephrine and dopamine, and most antidepressants are characterized by

interactions with these systems in the brain. More recently, new treatments for depression that

interact with non-monoamine systems have been studied (Hashimoto, 2009). The

glutamatergic system has been linked to MDD’s pathophysiology in many studies (Hasler et

al., 2007; Hashimoto, 2009; Zarate et al., 2010; Sanacora et al., 2012). This might explain the

lack of remission of depressive symptoms, which occurs in two-third of patients, with

medications that target only monoamine systems (Trivedi et al., 2006).

Ketamine, a noncompetitive antagonist of the glutamate receptor NMDA, is approved in New

Zealand as a general anesthetic (Ketalar® - Medsafe). After been used in Vietnam War, its

use was very extensive in the 1970’s, with the advantage of rarely causing respiratory

depression (Bonanno, 2002). Its applicability is also prominent for analgesia, being widely

used in clinical practice (Sih et al., 2011), however it is not approved in New Zealand for this

indication.

Ketamine was first studied as a possible antidepressant medication in 1975, when it was

compared to imipramine and placebo in rats, and showed positive results with doses of 40 and

80 mg/kg. In this study, ketamine was less effective than imipramine, however it was

significantly more effective than placebo (Sofia and Harakal, 1975). Importantly, this research

presented ketamine as possible treatment for depression. In 2000 the first clinical trial

evaluating ketamine as an antidepressant agent identified a rapid and sustained response, with

depression scores being reduced up to 3 days after a single IV infusion (Berman et al., 2000).

Since then, several studies have addressed the same objective, usually with similar outcomes

(Zarate et al., 2006; aan het Rot et al., 2010).

! -)!

In this review and meta-analysis, changes in depression ratings after dosing, from case reports

and randomized clinical trials, will be analyzed separately. The purpose of this analysis is to

determine the effectiveness, speed of onset, and duration of response, after a single infusion

of ketamine in patients with MDD.

5.2 Methods

5.2.1 Search strategy

Search was made in Pubmed, PsychInfo, Embase and ClinicalTrials.gov with the use of the

keywords “Ketamine” AND “depression”, “bipolar disorder”, OR “mood disorder”. No limits

were added, but only articles in English, Portuguese, Spanish and Italian were reviewed. The

search was made from any 1957 until present (November 2011). Also, citations in screened

articles were checked.

5.2.2 Selection criteria

Studies were required to meet the following criteria:

• patients with a diagnosis of MDD or bipolar disorder (BD) in a depressive episode

according to DSM criteria (DSM III, III-R, or IV, American Psychiatric Association,

1980, 1987, 2000),

• presenting moderate or severe depressive symptoms,

• use of ketamine with IV infusion over 40 to 60 minutes,

• dosage of 0.5mg/kg for racemic ketamine, or 0.25 mg/kg for S-ketamine.

Antidepressant response had to be assessed with reliable validated scales, such as

Mongomery-Asberg Depression Rating Scale (MADRS), Hamilton Rating Scale for

Depression (HAMD), Beck Depression Inventory (BDI), Hospital Anxiety and Depression

Scale (HADS) and/or The Quick Inventory of Depressive Symptomatology (QIDS-SR16).

Studies were analyzed to reach a consensus on what variables could be accepted within the

studies. Thus, patients with no history of ketamine treatment, and patients who had prior

ketamine use (for any treatment) could be included in review and meta-analysis. Current

medication could be suspended or maintained. Four studies used ketamine as an induction

agent for patients with MDD who were treated with electroconvulsive therapy (ECT). These

were not included in the review, as the use of ECT was a complicating factor.

! .+!

Data were included from case series and placebo-controlled randomized clinical trials

(RCTs).

Figure 5.1: Selection of studies for review and meta-analysis for ketamine as a fast acting

antidepressant.

Search of electronic databases:

35- ClinicalTrials.gov (16 selected) 0- Embase 725- Pubmed (36 selected) 8- PsychInfo (0 selected)

!!!!!!!!!!

1 article identified by other

search instruments (not selected)

54 articles were screened

39 articles excluded: 12 did not meet criteria 23 without enough data 3 overlapping studies 1 duplicated

12 case reports/ case series

included in review

3 RCTs included in

meta-analysis

! .%!

5.2.3 Analyses

RCTs were analyzed using RevMan 5.1(Cochrane Collaboration, 2008). Placebo versus

treatment were compared as an intervention review, with continuous data, random effects as

the analysis model and mean difference as effect measure along with 95% confidence

intervals.

Case reports and case series were compared with results from one of the RCTs. The choice of

using Diazgranados et al. data for this comparison was due to this study reporting changes in

three different depression scales. In total, 12 case reports/series results were compared to the

abovementioned RCT. Most of them (7) utilized MADRS for depressive symptoms

assessment, with a minority using HAMD and BDI.

5.3 Results

Fifteen studies were selected for comparison and review. All were published, with the first

one published in 2000 (Berman et al., 2000). There were 3 RCTs that analyzed the outcome

for ketamine as a fast-acting antidepressant.

5.3.1 Randomized clinical trials

Zarate et al. presented a crossover study design with 18 patients, and Diazgranados et al. used

a very similar study design 4 years latter. The primary diagnosis was the main difference

between these studies, as the first included only patients with MDD and the second, patients

with bipolar depression (BD).

Demographic and clinical characteristics of RCTs are presented on Table 5.1, where time to

response and time to relapse are clearly different when comparing the two most recent trials,

although the percentage of response and remission are very similar, 71% and 30%,

respectively. Unfortunately, the Berman et al., 2000 did not provide enough data for this

comparison. A substantial difference while comparing Berman et al., 2000 with the

abovementioned studies is that one of the inclusion criteria was depressive episode, with no

distinction between a diagnosis of MDD or BD, which may lead to different outcomes for

these patients’ subgroups.

! .*!

MADRS and HAMD were used as standard scales for depressive symptoms, which led the

possibility of a comparison of these trials using HAMD. There is a clear significant response

to ketamine when compared to placebo, and its best response occurs within 1 to 3 days in

most patients. As Berman et al., 2000 did not report depressive symptoms scores after 3 days,

subsequent results from other studies are not presented in figure 5.2, but will be discussed.

All RCTs reported ketamine side effects, with special attention to dissociative and euphoric

symptoms, which disappeared within 110 minutes after infusion and were not correlated to

change in depressive symptoms. Also, changes in vital signs were not clinically significant

and did not require any intervention. Most prevalent reactions were: dissociation, dizziness,

and changes in blood pressure and pulse.

Rates of response and side effects after infusion were very similar in all three studies, and the

fastest mood improvement could be observed within 40 minutes after infusion (Diazgranados

et al., 2010). Significant response (reduction of 50% or more in depression scales) within 24

hours was met in all RCTs (Berman et al., 2000; Zarate et al., 2006; Diazgranados et al.,

2010) and it lasted from 3 up to 7 days in most patients, with some of these subjects (11.7% in

Zarate et al., 2006) maintaining response for at least 2 weeks. In general, ketamine was

effective for 50% of patients in Berman et al., 2000 and for 71% in the other two RCTs. Table 5.1: Demographic and clinical data for RCTs.

* 8 patients with recurrent MDD and 1 patient with BD – depressive episode. ** Patients were allowed to use lithium and/or valproate. *** First assessment after baseline.

Study Berman et al., 2000 (n=9) Zarate et al., 2006 (n=18)

Diazgranados et al., 2010 (n=18)

Depression scales used HDRS; BDI HDRS21; BDI HDRS17; BDI; MADRS

Age (years) 37 (± 10) 46.7 (± 11.2) 47.9 (± 13.1) Gender 55.5% female 66% female 67% female Diagnosis 88.9% recurrent MDD * Recurrent MDD BD - depressive

episode Medication wash-out yes yes yes ** Treatment resistant data not available yes yes Length of illness (years) data not available 23.7 (± 12.5) 27.6 (± 11.2) Length of current episode (months) data not available 33.6 (± 37.4) 15.1 (± 13.3) Time to significant improvement 240 minutes*** 110 minutes 40 minutes Time to relapse (days) 7 to 14 7 days 3 days Response during treatment (%) 50 71 71 Remission during treatment (%) 26 29 31

! .,!

Figure 5.2: HAMD mean values for Diazgranados et al., 2010, Zarate et al., 2006, and Berman et al., 2000. !!

!

10

15

20

25

30

35

baseline 40-60 min 230-240 min day 1 day 2 day 3

placebo - Diazgranados et al., 2010 ketamine - Diazgranados et al., 2010

placebo - Zarate et al., 2006 ketamine - Zarate et al., 2006

placebo - Berman et al., 2000 ketamine - Berman et al., 2000

! .-!

5.3.2 Case reports and case series

All patients had a diagnosis of Major Depressive Disorder, and most of them presented a

treatment resistant depressive episode, even though this information was not provided in one

study (Valentine et al., 2011) and was not applicable to one patient (Stefanczyk-Sapieha et al.,

2008).

In all cases, as in the RCTs, patients who responded to ketamine infusion did so within the

first 24 hours. In most of them, improvement in depressive symptoms could be noted in the

first 4 hours (Stefanczyk-Sapieha et al., 2008; Liebrenz et al., 2009; Machado-Vieira et al.,

2009; Phelps et al., 2009; Denk et al., 2011; Ibrahim et al., 2011; Salvadore et al., 2011;

Valentine et al., 2011). Time to relapse was not clear in some cases (Machado-Vieira et al.,

2009; Phelps et al., 2009; Denk et al., 2011; Ibrahim et al., 2011; Salvadore et al., 2011), but

when available they varied from 3 to 22 days, as summarized in Table 5.2.

Results from most cases corroborate to findings in RCTs. Figure 5.3 illustrates the analysis

between results from case reports/series and outcomes from Diazgranados et al., 2010 using

HAMD scale. In the first 168 hours, studies from Paul et al., 2009 and Liebrenz et al., 2009

more clearly illustrate the similar outcomes when compared to the RCT. This is due to the

timing of post-infusion assessments, which made the comparisons of ratings possible.

Antidepressant response to ketamine is also illustrated in Figure 5.4, with MADRS for

depressive ratings, and in Figure 5.5, with BDI as depression scale for assessments.

! ""!

Table 5.2: Demographic and clinical data for cases.

!

Study Caric et

al., 2007

(n=1)

Stefanczyk-

Sapieha et al.,

2008 (n=1)

Liebrenz

et al., 2009

(n=1)

Denk

et al., 2011

(n=1)

Paul

et al., 2009

(n=2)

Valentine

et al., 2011

(n=10)

Salvadore

et al., 2011

(n=14)

Machado-

Vieira et al.,

2009 (n=23)

Phelps et al.,

2009 (n=26)

Mathew

et al., 2010

(n=26)

Ibrahim

et al., 2011

(n=40)

Depression scales

used

BDI HDRS21; BDI BDI; HDRS21 MADRS;

BDI

BDI;

HDRS21

HDRS25 MADRS MADRS MADRS;

HDRS17;

BDI

MADRS;

QIDS-SR16

MADRS

Age (years) 32 50 55 56 54.5 41.7 (± 12) 50.1 (± 10.4) 43.9 (± 13.9) 43.5 (± 14.1) 48.2 (± 11.8) 46.5

Gender male male male female 50%

female

60%

female

35.8%

female

39%

female

39%

female

39%

female

40%

female

Diagnosis MDD MDD MDD MDD recurrent

MDD

MDD * MDD MDD MDD MDD MDD

Medication wash-out no no no NA no yes yes yes yes yes yes

Treatment resistant yes no yes yes yes NA yes yes yes yes yes

Length of illness

(years)

NA NA NA NA 7.5 21.2 (±

17.4)

28.9 (± 14.2) 24.8 (± 12.4) 23.7 (± 12.5) 29.7 25.3

Length of current

episode

NA NA NA NA NA 3.3 years

(± 4.3)

13.2 years

(± 15)

1.7 years

(± 2.26)

6.8 years

(± 8.6)

24.3 years

(± 16.3)

8.3 years

Time to significant

improvement

24 hours 2 hours 24 hours NA 24 hours 1 hour 4 hours 3.83 hours 3.83 hours 2 hours 3.83 hours

Time to relapse

(days)

10 3 7 NA 6 7 NA NA NA 22 NA

Response during

treatment

yes yes yes yes 50% yes, % NA yes, % NA 47.8% 43% 73% 50%

Remission during

treatment

yes no not clear yes no NA NA NA 26% 50% NA

NA = data not available Not clear = meets criteria for 1 assessed scale but not for the second applied scale !!!!!#$%&!'()*''(+,!!

! "#!

Figure 5.3: Cases outcomes and Diazgranados et at., 2010 (HAMD).

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!! !

Ketamine effect on depression in case reports and case series over time according to changes in HAMD ratings. Graphs (B) and (C) show changes over shorter time intervals.!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

$%&'!()!%&*+!,%-%*.%/!,.0/1!)(2!34$5!*(67&28,('!9:;!<#=!+>!

$%&'!()!%&*+!,%-%*.%/!,.0/1!)(2!34$5!*(67&28,('!9:;!?@!+>!

$%&'!()!%&*+!,%-%*.%/!,.0/1!)(2!34$5!*(67&28,('!9:;!#!+>!!"#

$"#

%"#

! "A!

Figure 5.4: Cases outcomes and Diazgranados et at., 2010 (MADRS).

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!!!!!!!!!!!!!!!!!!!! !

Ketamine effect on depression in case reports and case series over time according to changes in MADRS ratings. Graph (B) shows changes over a shorter time interval.

!

!

!%"!!!!!!!!$%&'!()!%&*+!,%-%*.%/!,.0/1!)(2!$45BC!*(67&28,('!9:;!?@!+>!!

!

$"!!!!!!!!$%&'!()!%&*+!,%-%*.%/!,.0/1!)(2!$45BC!*(67&28,('!9:;!@!+>!!

!

! "=!

Figure 5.5: Case outcome and Diazgranados et at., 2010 (BDI).

!

!

!

0 10 20 30 40

"#!

$#!

%#!

&#!

#!

'#!

0 10 20 30 40

@:!

?:!

<:!

D:!

:!

":!

!

0 10 20 30 40

"#!

$#!

%#!

&#!

#!

'#!

!

0 10 20 30 40

"#!

$#!

%#!

&#!

#!

'#!

!

0 10 20 30 40

"#!

$#!

%#!

&#!

#!

'#!

!

0 10 20 30 40

"#!

$#!

%#!

&#!

#!

'#!

!

0 10 20 30 40

"#!

$#!

%#!

&#!

#!

'#!

!

0 10 20 30 40

"#!

$#!

%#!

&#!

#!

'#!

Mean of selected study for comparison

! "E!

5.3.3 Meta-analysis

In all selected RCTs, a significant antidepressant response was found within 230- 240 minutes

post-infusion. Also, this response was sustained for at least 3 days. HAMD ratings were

relatively stable after 230- 240 minutes post-infusion in the outcomes from Berman et al.,

2000 and Diazgranados et al., 2010. However, Zarate et al., 2006 had a continuous decrease

in depression ratings until day 3 post-infusion (Figure 5.2).

Although Diazgranados et al., 2010 presents a significant treatment effect at 40 minutes after

ketamine infusion, the two other RCTs do not have a significant response within this time.

Thus, the test for overall effect in the meta-analysis results was not statistically significant

(Figure 5.6 A). Diazgranados et al., 2010 presented a faster response than other RCTs and a

shorter duration of ketamine’s antidepressant effects. The possible reasons for this difference

only in Diazgranados et al., 2010 will be discussed further.

At 230 – 240 minutes post-infusion, the three studies are homogeneous and the change in

HAMD scores compared with placebo are substantial and highly statistically significant (5.6

B). At days 1, 2, and 3 post-infusion there are also substantial reductions in HAMD scores

(Figures 5.6 C, D, E). Heterogeneity was evident at days 1, 2, and 3 (highest I2 scores), which

was due to data from Diazgranados et al., 2010.

! #:!

Figure 5.6: Meta-analysis results based on change in HAMD scores.

Study or SubgroupBerman, 2000Zarate, 2006Diazgranados, 2010

Total (95% CI)Heterogeneity: Tau! = 1.29; Chi! = 2.73, df = 2 (P = 0.25); I! = 27%Test for overall effect: Z = 1.75 (P = 0.08)

Mean-1.6-5.2

-6.47

SD6.45.3

4.05

Total8

1717

42

Mean-1.5

-4.28-2.54

SD1.556.253.96

Total8

1716

41

Weight23.0%29.4%47.6%

100.0%

IV, Random, 95% CI-0.10 [-4.66, 4.46]-0.92 [-4.82, 2.98]

-3.93 [-6.66, -1.20]

-2.17 [-4.60, 0.27]

Year200020062010

ketamine placebo Mean Difference Mean DifferenceIV, Random, 95% CI

-20 -10 0 10 20Favours experimental Favours control

Study or SubgroupBerman, 2000Zarate, 2006Diazgranados, 2010

Total (95% CI)Heterogeneity: Tau! = 0.00; Chi! = 0.13, df = 2 (P = 0.93); I! = 0%Test for overall effect: Z = 5.60 (P < 0.00001)

Mean-8

-11.42-8.83

SD8

5.53.96

Total8

1717

42

Mean-1.8

-4.98-3.24

SD1.3

6.1253.96

Total8

1716

41

Weight13.6%27.9%58.5%

100.0%

IV, Random, 95% CI-6.20 [-11.82, -0.58]-6.44 [-10.35, -2.53]-5.59 [-8.29, -2.89]

-5.91 [-7.98, -3.84]

Year200020062010

ketamine placebo Mean Difference Mean DifferenceIV, Random, 95% CI

-20 -10 0 10 20Favours experimental Favours control

Study or SubgroupBerman, 2000Zarate, 2006Diazgranados, 2010

Total (95% CI)Heterogeneity: Tau! = 15.67; Chi! = 9.57, df = 2 (P = 0.008); I! = 79%Test for overall effect: Z = 3.44 (P = 0.0006)

Mean-11.1

-13.72-7.36

SD8.8

53.9

Total8

1717

42

Mean1.5

-2.68-2.54

SD3.3

6.253.9

Total8

1716

41

Weight25.6%35.2%39.1%

100.0%

IV, Random, 95% CI-12.60 [-19.11, -6.09]-11.04 [-14.84, -7.24]

-4.82 [-7.48, -2.16]

-9.01 [-14.14, -3.88]

Year200020062010

ketamine placebo Mean Difference Mean DifferenceIV, Random, 95% CI

-20 -10 0 10 20Favours experimental Favours control

Study or SubgroupBerman, 2000Zarate, 2006Diazgranados, 2010

Total (95% CI)Heterogeneity: Tau! = 1.71; Chi! = 2.76, df = 2 (P = 0.25); I! = 28%Test for overall effect: Z = 5.90 (P < 0.00001)

Mean-12.8

-12.05-8.59

SD85

3.96

Total8

1717

42

Mean-0.8-2.5

-2.06

SD7.9

6.1254.08

Total8

1716

41

Weight11.1%36.0%52.9%

100.0%

IV, Random, 95% CI-12.00 [-19.79, -4.21]-9.55 [-13.31, -5.79]-6.53 [-9.28, -3.78]

-8.22 [-10.96, -5.49]

Year200020062010

ketamine placebo Mean Difference Mean DifferenceIV, Random, 95% CI

-20 -10 0 10 20Favours experimental Favours control

Study or SubgroupBerman, 2000Zarate, 2006Diazgranados, 2010

Total (95% CI)Heterogeneity: Tau! = 7.45; Chi! = 5.21, df = 2 (P = 0.07); I! = 62%Test for overall effect: Z = 3.79 (P = 0.0001)

Mean-13.2

-11.22-6.94

SD8

5.1254.08

Total8

1717

42

Mean-0.2-2.5

-2.06

SD7.9

6.254.08

Total8

1716

41

Weight18.1%37.3%44.5%

100.0%

IV, Random, 95% CI-13.00 [-20.79, -5.21]-8.72 [-12.56, -4.88]-4.88 [-7.67, -2.09]

-7.79 [-11.81, -3.76]

Year200020062010

ketamine placebo Mean Difference Mean DifferenceIV, Random, 95% CI

-20 -10 0 10 20Favours experimental Favours control

A) Antidepressant response 40 minutes post-infusion.

B) Antidepressant response 230 – 240 minutes post-infusion.

C) Antidepressant response 1 day post-infusion.

D) Antidepressant response 2 days post-infusion.

E) Antidepressant response 2 days post-infusion.

! #<!

5.4 Discussion

Several RCTs and multiple case reports/series have consistently showed ketamine to be a

rapidly acting antidepressant. Researches with ketamine as a fast acting antidepressant have

not been limited to intravenous administration; some authors have explored alternative routes,

such oral (Irwin and Iglewicz., 2010; Paslaskis et al., 2010) and intramuscular routes (Glue et

al., 2011). The optimal dose and whether it can be used repeatedly, is also not clear. For

example, 0.2mg/kg of ketamine in bolus has demonstrated to be effective for suicidal ideation

(Larkin and Beautrais, 2011). Even more, another study has contributed to these findings,

with promising results for suicidal ideation in treatment resistant MDD, with 0.5mg/kg of

ketamine administered over 40 minutes intravenously (Price et al., 2009). To evaluate dose

response, Glue et al., 2011 compared results for the same patients (n=2) when receiving 0.5,

0.7, and 1mg/kg of intramuscular ketamine. A better outcome was found with the dosage of

1mg/kg, leading us to the question: Would patients, who did not respond to ketamine in other

studies, have a significant response with higher doses? Further investigation is required to

establish how this possible treatment could get to its best results.

In this review, S (+)-ketamine 0.25mg/kg was considered for comparison with racemic

ketamine 0.5mg/kg, as the first is considered to be twice as potent as the racemic mixture

(Hering et al., 1994). According to some studies, S(+) and racemic ketamine do not differ

significantly on hemodynamic changes (White et al., 1985; Ihmsen et al., 2001), being as safe

as racemic ketamine.

The studies analyzed included treatment resistant patients, and their positive outcomes may

indicate an option for this population. Although there were differences in patients’ response

when analyzing MDD and depressive episode in BD (possibly due to the different diagnoses,

use of lithium and valproate in patients with BD, and/or severity of illness) both groups had

generally similar mood responses (Zarate et al., 2006; Diazgranados et al., 2010). Group with

BD may possibly have had faster responses but shorter durations of positive effects

(Diazgranados et al., 2010). This requires future study A similar methodology with a

complete medication washout could exclude the medication factor for analyses. In all studies,

difficulty in maintaining blinding of groups and researchers was clear, as ketamine side

effects (such as euphoria and depersonalization) were easily recognizable. Also, in all RCTs,

depersonalization was not correlated to change in depressive symptoms, indicating that

psychomimetic effects are not required to a positive result.

! #?!

Continuous infusions (with the same methodology used for the first infusion) were present in

3 of the reviewed cases. Stefanczyk-Sapieha et al., 2011 presented a patient who had a shorter

response (24 hours) to ketamine after a second infusion, while the first infusion provided

depression response for up to 3 days. By the other hand, Paul et al., 2009 and Liebrenz et al.,

2009 presented outcomes in a second infusion that were similar to the first ketamine

administration.

Types of rating scales, and timing of ratings, varied across reports. The MADRS and HAMD

were the most commonly used depression scales. Ideally, time points from baseline to 40

minutes, 120 minutes, 240 minutes, 1 day, 3 days, 7 days and more days if response is

maintained may provide important observations for future reviews, as this information was

not reported in many studies. Results were consistent between 4 hours and 3 days post-

infusion, and may be of important consideration for future studies.

5.5 Conclusions

Ketamine has been extensively studied as an antidepressant in the last decade. There are

promising data in terms of rapidity of mood improvement in RCTs involving patients with

refractory unipolar and bipolar depression. Data from case reports and case series are

generally similar to those reported in the RCTs. Significant areas that require clarification

include what the optimal dose and route of administration are, and whether ketamine can be

used in patients with less refractory mood disorders. Although mood responses in unipolar

and bipolar depression appear to be similar, it may be prudent to only include one or other

patient group in future RCTs, and not to combine them. Methodologically, it would be

important for investigators to use similar rating scales and time-points, to maximize

comparisons between studies.

! #D!

CHAPTER 6

Protocol for testing ketamine in Major

Depressive Disorder in terminal cancer

patients

! #@!

6.1 Introduction

This protocol was based on studies that present positive results of ketamine as a fast acting

antidepressant (Diazgranados et al., 2010; Zarate et al, 2006; Berman et al., 2000).

Considering that patients with terminal cancer have a short life expectancy and high rates of

depression, ketamine may be a possible treatment for this population. This trial has been

approved by the Lower South Regional Ethics Committee and is a registered trial -ACTRN

12610001011077. It is one of the first trials of ketamine for depression in patients with

cancer. In particular, this group of patients may benefit from the analgesic effects of

ketamine, as pain rates are also high in this population. Its design addresses a number of key

clinical questions in a scientifically rigorous manner, including:

1- Confirmation that low dose ketamine is an effective antidepressant in a palliative care

cancer population;

2- Demonstration that repeated doses are safe and effective;

3- Characterization of dose-response profile, particularly as this patient group may be

physically debilitated.

The trial consists of 3 stages, each one concerning to the abovementioned objectives, and all

stages take place in a hospital room, with emergency support if needed.

6.2 Stages

6.2.1 Stage 1

Open label characterization of safety and efficacy of 1 mg/kg single dose IM ketamine in 10

depressed patients with cancer. If an antidepressant response can be demonstrated in 5/10

patients, with tolerable side effects, subjects are eligible to proceed to stages 2 and 3. A 50%

improvement in depressive symptoms in MADRS is considered response to treatment

(Zimmerman et al., 2004; Zarate et al., 2006).

6.2.2 Stage 2

Open label characterization of safety and efficacy of 1mg/kg repeat dose IM ketamine in 10

depressed patients with cancer. Subjects that present a sustained response, with tolerable side

effects, are eligible to keep receiving treatment if symptoms recurred. Patients had the option

to drop out of the trial at any time.

! #"!

6.2.3 Stage 3

Double blind, randomized, dose-response assessment of single dose IM ketamine in 30

depressed patients with cancer. Injection dose might be 0.1mg/kg, 0.5mg/kg or 1mg/kg.

Subjects can decide not to participate on Stage 3 at any time.

6.3 Inclusion and exclusion criteria

6.3.1 Inclusion criteria:

• ECOG performance status ! 3 (Table 6.1),

• MADRS >20 (Figure 6.1),

• Comparative Pain Scale ratings ! 5 (Table 6.2),

• presence of terminal (incurable) cancer,

• life expectancy of at least 3 months,

• aged 20 to 65 years-old,

• adequate hematological, renal and hepatic function,

• signed informed consent.

MADRS scores >20 are standard for entry into antidepressant clinical trials (Machado-Vieira

et al., 2009; Diazgranados et al., 2010). Patients with mild to moderate levels of pain were

eligible, but patients with severe pain were excluded, as there might be a bias in patients’

outcomes (as a response in depressive symptoms might be due to pain relief). Subjects could

be confined to bed or chair more than 50% of waking hours and with limited self-care, but not

totally confined to bed or chair or completely disabled, as this would limit data collection.

Further, considering the risk of arrhythmias, slow urinary excretion, and slow hepatic

metabolism, cardiac, renal and hepatic function had to be adequate. Also, patients had to

provide written informed consent.

6.3.2 Exclusion criteria:

• presence of psychosis or delirium,

• presence of cerebral metastasis,

• current use of ketamine,

• current use of another experimental medication,

• any antidepressant treatment started within the last 4 weeks.

! ##!

Considering psychosis and delirium, as there is a lack of reasoning and judgment, patients

would not be able to give informed consent, and the use of ketamine in these population,

might cause a worsening in their symptoms (Ketalar- Medsafe). The presence of cerebral

metastasis may cause neurologic and psychiatric symptoms and change the permeability of

the blood-brain barrier (Chakrabarti et al., 2011; Lee 2010; Abbott et al., 2006). The last

might lead to a different dosage of the medication acting in the brain. Current use of ketamine

was not allowed because it might be already impacting patient’s mood, and experimental

medication because of possible unknown influence in depressive symptoms. Subjects could

not be receiving any new treatment for depression in the last 4 weeks (including

psychotherapy), to avoid positive results that might not be linked to ketamine administration.

Entry criteria assessment sheets can be found in attachments.

Table 6.1: ECOG performance status.

Table from: Oken et al., 1982.

Grade ECOG 0 Fully active, able to carry on all pre-disease performance without restriction 1 Restricted in physically strenuous activity but ambulatory and able to carry out

work of a light or sedentary nature, e.g., light house work, office work 2 Ambulatory and capable of all selfcare but unable to carry out any work

activities. Up and about more than 50% of waking hours 3 Capable of only limited selfcare, confined to bed or chair more than 50% of

waking hours 4 Completely disabled. Cannot carry on any selfcare. Totally confined to bed or

chair 5 Dead

! #A!

Figure 6.1: MADRS.

! #=!

Table 6.2: The Comparative Pain Scale.

0

No pain. Feeling perfectly normal.

Minor

Does not interfere with most activities. Able to adapt to pain psychologically and with medication or devices such as cushions.

1

Very Mild

Very light barely noticeable pain, like a mosquito bite or a poison ivy itch. Most of the time you never think about the pain.

2

Discomforting

Minor pain, like lightly pinching the fold of skin between the thumb and first finger with the other hand, using the fingernails. Note that people react differently to this self-test.

3 Tolerable

Very noticeable pain, like an accidental cut, a blow to the nose causing a bloody nose, or a doctor giving you an injection. The pain is not so strong that you cannot get used to it. Eventually, most of the time you don't notice the pain. You have adapted to it.

Moderate

Interferes with many activities. Requires lifestyle changes but patient remains independent. Unable to adapt to pain.

4 Distressing

Strong, deep pain, like an average toothache, the initial pain from a bee sting, or minor trauma to part of the body, such as stubbing your toe real hard. So strong you notice the pain all the time and cannot completely adapt. This pain level can be simulated by pinching the fold of skin between the thumb and first finger with the other hand, using the fingernails, and squeezing real hard. Note how the simulated pain is initially piercing but becomes dull after that.

5

Very Distressing

Strong, deep, piercing pain, such as a sprained ankle when you stand on it wrong, or mild back pain. Not only do you notice the pain all the time, you are now so preoccupied with managing it that you normal lifestyle is curtailed. Temporary personality disorders are frequent.

6 Intense

Strong, deep, piercing pain so strong it seems to partially dominate your senses, causing you to think somewhat unclearly. At this point you begin to have trouble holding a job or maintaining normal social relationships. Comparable to a bad non-migraine headache combined with several bee stings, or a bad back pain.

Severe

Unable to engage in normal activities. Patient is disabled and unable to function independently.

7

Very Intense

Same as 6 except the pain completely dominates your senses, causing you to think unclearly about half the time. At this point you are effectively disabled and frequently cannot live alone. Comparable to an average migraine headache.

8

Utterly Horrible

Pain so intense you can no longer think clearly at all, and have often undergone severe personality change if the pain has been present for a long time. Suicide is frequently contemplated and sometimes tried. Comparable to childbirth or a real bad migraine headache.

9

Excruciating Unbearable

Pain so intense you cannot tolerate it and demand pain killers or surgery, no matter what the side effects or risk. If this doesn't work, suicide is frequent since there is no more joy in life whatsoever. Comparable to throat cancer.

10

Unimaginable Unspeakable

Pain so intense you will go unconscious shortly. Most people have never experienced this level of pain. Those who have suffered a severe accident, such as a crushed hand, and lost consciousness as a result of the pain and not blood loss, have experienced level 10.!

! #E!

6.4 Informed consent

For each subject, a consent form, which was written in lay terms, was given and thoroughly

explained. After having complete understanding of the trial, patients had up to 2 days to think,

talk to friends, family, other doctors, etc. Further, individuals were reassured that they could

withdraw their consent at anytime, without needing to explain or giving a reason for it. It was

also emphasized that withdrawing would not affect their health treatment in any way. A copy

of the informed consent is attached to this thesis (Appendix I).

6.5 Assessments

In all three stages, two main scales were used to access depression ratings (MADRS; Figure

6.1) and pain (The Comparative Pain Scale; Table 6.2). These were performed at screening

(20 minutes before injection) and 1, 2, 4, 24, 48, 72, and 120 hours post-injection. Other

scales used in the study were: Hospital Anxiety and Depression Scale (HADS; Figure 6.2)

and Demoralization Scale (Figure 6.3), which were performed at baseline and 24 hours post-

injection.

MADRS is a well known and validated scale to access depression (Phelps et al., 2009;

Diazgranados et al., 2010). Also, it includes all DSM-IV criteria for MDD (Montgomery and

Asberg, 1979). Its main difference when compared to HADS is that the last one does not

include physical symptoms in its rating. Therefore, HADS is often used in patients with

cancer and other somatic diseases (Zigmond and Snaith, 1983; Bjelland et al., 2002).

Demoralization Scale consists of 24 questions with a total score ranging from 0 to 96, and a

score > 30 is considered high. This suggests the presence of adjustment disorder or even a

depressive episode (Kissane et al., 2004). Considering the fact that cancer patients usually

experience pain, Comparative Pain Scale was used to assess its intensity. This scale consists

of scores that range from 0 to 10, considering 0 as “no pain” and 10 as “unspeakable pain”.

Also, it is going to be useful to check correlation between pain and depression, as many

studies report their high correlation, even though its neural basis is not yet elucidated (Bair et

al., 2003; Strigo et al., 2008).

Vital signs were assessed on baseline, 1, 2, 3, and 4 hours post-injection on stage 1 and 3. At

stage 2, if patients maintain a stable and normal blood pressure, after 3 repeat dosing,

assessments are made on baseline, 1, and 2 hours post-injection. Vital signs assessments are

repeated at any time if patients present distressing side effects or report possible cardiac

! A:!

symptoms, such as tachycardia, palpitation, tachypnoea or bradypnea, feeling of syncope, etc.

Side effects are monitored all the time, for up to 2 or 4 hours, depending on the stage in which

patients are.

Figure 6.2: HADS.

! A<!

Figure 6.3: The Demoralization Scale. F(2!%&*+!,.&.%6%'.!G%-(HI!1(0!&2%!&,J%/!.(!8'/8*&.%!+(H!,.2('K-1!.+%!,.&.%6%'.!+&,!&77-8%/!.(!1(0!(L%2!

.+%!-&,.!.H(!H%%J,!G1!*82*-8'K!.+%!*(22%,7('/8'K!'06G%2M!NL%2!.+%!7&,.!.H(!H%%J,!+(H!().%'!+&L%!1(0!

)%-.O!

# &'(')# *'+,-.# *-.'/01.'2# 340'5# %++#06'#

71.'#

<M!!!P+%2%!8,!&!-(.!()!L&-0%!8'!H+&.!Q!*&'!())%2!

(.+%2,M!:! <! ?! D! @!

?M!!!!$1!-8)%!,%%6,!.(!G%!7(8'.-%,,M! :! <! ?! D! @!

DM!!!!P+%2%!8,!'(!7027(,%!.(!.+%!&*.8L8.8%,!8'!61!-8)%M!! :! <! ?! D! @!

@M!!!!$1!2(-%!8'!-8)%!+&,!G%%'!-(,.M! :! <! ?! D! @!

"M!!!!Q!'(!-('K%2!)%%-!%6(.8('&--1!8'!*('.2(-M! :! <! ?! D! @!

#M!!!!Q!&6!8'!K((/!,7828.,M! :! <! ?! D! @!

AM!!!!R(!('%!*&'!+%-7!6%M!!! :! <! ?! D! @!

=M!!!!Q!)%%-!.+&.!Q!*&''(.!+%-7!61,%-)M! :! <! ?! D! @!

EM!!!!Q!)%%-!+(7%-%,,M! :! <! ?! D! @!

<:M!!Q!)%%-!K08-.1M! :! <! ?! D! @!

<<M!!Q!)%%-!8228.&G-%M! :! <! ?! D! @!

<?M!!Q!*(7%!)&82-1!H%--!H8.+!-8)%M! :! <! ?! D! @!

<DM!!Q!+&L%!&!-(.!()!2%K2%.!&G(0.!61!-8)%M! :! <! ?! D! @!

<@M!!S8)%!8,!'(!-('K%2!H(2.+!-8L8'KM!!! :! <! ?! D! @!

<"M!!Q!.%'/!.(!)%%-!+02.!%&,8-1M! :! <! ?! D! @!

<#M!!Q!&6!&'K21!&G(0.!&!-(.!()!.+8'K,M! :! <! ?! D! @!

<AM!!Q!&6!72(0/!()!61!&**(67-8,+6%'.,M! :! <! ?! D! @!

<=M!!Q!)%%-!/8,.2%,,%/!&G(0.!H+&.!8,!+&77%'8'K!.(!

6%M!:! <! ?! D! @!

<EM!!Q!&6!&!H(2.+H+8-%!7%2,('M! :! <! ?! D! @!

?:M!!Q!H(0-/!2&.+%2!'(.!G%!&-8L%M! :! <! ?! D! @!

?<M!!Q!)%%-!,&/!&'/!68,%2&G-%M! :! <! ?! D! @!

??M!!Q!)%%-!/8,*(02&K%/!&G(0.!-8)%M! :! <! ?! D! @!

?DM!!Q!)%%-!T08.%!8,(-&.%/!(2!&-('%M! :! <! ?! D! @!

?@M!!Q!)%%-!.2&77%/!G1!H+&.!8,!+&77%'8'K!.(!6%M! :! <! ?! D! @!

!"#$%&'()*&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&&R(.%!H+%'!,*(28'KU!8.%6,!<I!#I!<?I!<A!&'/!<E!&2%!2%L%2,%!,*(2%/M&

! A?!

6.6 Statistical Analysis

All data were listed, and summary statistic (mean, SD) calculated. Change scoring in MADRS

and pain ratings were calculated by subtracting scores post-dosing from baseline values.

Correlation of change in pain and depression scores was made using Pearson’s method.

6.7 Conclusions

This protocol presents similarities with previous studies, however it adds pain scores.

Ketamine has analgesic properties, thus correlation between pain and depression responses

can be assessed. Results found with this methodology may shed light to the relationship

between pain and depression as they are frequently coexistent and some authors claim their

interdependence (Mongini et al., 2007; Waters et al., 2004). Also, another aspect of this

protocol is the possibility of repeated ketamine injections for Major Depression.

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

! AD!

CHAPTER 7

Acute and maintenance IM ketamine for a

depressed patient with terminal cancer

! A@!

7.1 Introduction

This chapter describes a patient enrolled in a clinical trial to assess the effects of Ketamine in

MDD in patients with terminal cancer. The trial was approved by the Lower South Regional

Ethics Committee, Southern Health District Board and Dunedin School of Medicine,

University of Otago, New Zealand (ACTRN 12610001011077). The patient participated in

Stages 1 and 2 of this trial.

7.2 Case Report

A. is a 37-year-old patient with metastatic ovarian carcinoma. She has a diagnosis of Major

Depressive Disorder (MDD) and Dysthymic Disorder since she was 18 years old. Although

she has used various antidepressants, A. reports her mood was never fully improved. For the

past 10 years she presented a worsening of her depressive symptoms, which persisted at the

time of her entrance in the study, including: irritability, low mood, lack of energy, easy

crying, sleep disturbance, poor concentration, intense and excessive feelings of guilt and

suicidal ideation. Her domestic circumstances were very stressful, due to concern about the

future of her 11-year-old daughter and the lack of support from her friends and family.

The most distressing symptoms of her metastatic ovarian carcinoma, was visceral pain in the

right iliac fossa, which started 4 months after her diagnosis of cancer. Her diagnosis was

made in January of 2010, and her pain was in the same location as a metastatic tumor, which

was about 7cm in diameter on palpation in November of 2011. For pain intensity assessment,

The Comparative Pain Scale, with a range from 0 to 10 was used (Harich- online resource).

She also has a previous history of drug abuse, which started when she was 17 years old, with

the use of cannabis. At 21 years old, she used cocaine, heroin and methamphetamine. With

time, her drug of choice was methamphetamine. In 2004 she was able to quit the use of drugs

after starting methadone for opioid substitution therapy. A. has been abstinent since then.

There is also a family history of alcoholism (both grandfathers and one uncle).

When first assessed for this ketamine trial, A. was taking venlafaxine 150 mg/day, quetiapine

50 mg/day, and methadone 95 mg/day. All her medications were continued during her

ketamine treatment with doses unchanged. She was also receiving chemotherapy once a week

(a combination of carboplatin and gemcitabine). After informed consent was given, she

received ketamine 1 mg/kg IM open-label, and her dosing was performed on an outpatient

basis in a hospital clinic.

! A"!

The Montgomery-Asberg Scale (MADRS) was used as the main scale to assess her

depression severity. This scale consists of 10 items, each one scored from 0 to 6 (total range

from 0 to 60). Scores > 20 are standard for entry into antidepressant clinical trials

(Diazgranados et al., 2010; Machado-Vieira et al., 2009). Mood and pain assessments were

obtained at baseline (20 minutes before injection), 1, 2, 4, 24, 48, 72 and 168 hours post-

injection. The Hospital Anxiety and Depression Scale (HADS) is often used in patients with

cancer or other somatic diseases, and its optimal cut off scores are > 7 on both HADS-A and

HADS-D for anxiety and depression respectively (Zigmond and Snaith, 1983; Bjelland et al.,

2002). No physical symptoms are included in this scale, permitting a focus on affective and

behavioral symptoms (Zigmond and Snaith, 1983). The Demoralization Scale (DS) consists

of 24 questions (with a total score range from 0 to 96), and a score > 30 is considered high

(Kissane et al., 2004). HADS and DS were assessed at baseline and 24 hours.

The patient’s initial MADRS score was 24, dropping to 7 one hour after the first dose of

ketamine (Figure 7.1 A). For the next 2 days her MADRS remained low (lowest score was 5

points), with improvement in all MADRS items. Depressive symptoms returned after 6 - 7

days post-injection. Also, her pain score was reduced within 15 to 20 minutes after the first

ketamine dose, however her scores for pain returned to baseline within 24 hours (Figure 7.1

A). A. reported moderate but tolerable dissociative side effects, such as depersonalization and

immobility, starting 5 minutes post-injection and resolving within 45 to 60 minutes. Vital

signs were unchanged.

The patient subsequently received repeat ketamine doses at intervals of 8 to 10 days, based on

return of her depressive symptoms, initially receiving treatment when her depressive scores

were higher than 20. Her mood response to repeat injections has been identical each time, in

terms of speed, magnitude of response and duration (Figure 7.1 B). Onset of pain relief

continued to be fast, nevertheless the durability of analgesic response was much more variable

(Figure 7.1 C). Reported side effects were similar in timing of onset and intensity to those

noted after the first dose of ketamine. Scores on HADS and DS demonstrated consistent

improvement in her mood symptoms (Figure 7.2). Correlation between changes in depression

and pain scores over time were initially robust, but decreased over the next 168 hours (Figure

7.3).

! A#!

Figure 7.1: Antidepressant and analgesic response to IM ketamine 1mg/kg.

!

A) Antidepressant and analgesic effects after first IM injection of ketamine 1 mg/kg – Stage 1. B) Consistency of antidepressant response after 6 injections. C) Inconsistency in pain response after 24 hours after 6 injections.

V&8'!9!!>!

:!!!!!!!!!!!!!?@!!!!!!!!!!!!@=!!!!!!!!!!!!A?!!!!!!!!!!!!!E#!!!!!!!!!!!<?:!!!!!!!!!<@@!!!!!!!!!!<#=!!!!!!!!!!<E?!

!

?"!

?:!

D:!

<:!

!!"!

!:!

<"!

D!

?!

<!

:!

@!

"!

$45BC!9!!>!

V&8'!2&.8'K!9!!>!

.86%!9+>!!

!!$45BC!

!!!!!.86%!9+>!

:!!!!!!!!!!!!!?@!!!!!!!!!!!!@=!!!!!!!!!!!!A?!!!!!!!!!!!!!E#!!!!!!!!!!!<?:!!!!!!!!!<@@!!!!!!!!!!<#=!!!!!!!!!!<E?!

?"!

?:!

D:!

<:!

!!"!

!:!

<"!

$45BC!

time (h) 0 24 48 72 96

!

120 144 168 192

V&8'!2&.8'K!

0

1 !

2

3

4

5

6

7

! !

!

! !

!

!

! !

!

! ! ! ! !

! !

! ! !

!

!

! ! !

!

!

!

!

! !

! !

!

!

! !

! ! ! !

! !

! !

! ! !

!

!

! !

!

! !

! Dose 1 Dose 2 Dose 3 Dose 4 Dose 5 Dose 6

! ! ! ! ! !

!!

!

!!

!

!

!!

!

! ! ! !!

!!

! ! !

!

!

!!!

!

!

!

!

!!

! !

!

!

!!

!!! !

! !

!!

!!!

!

!

!!

!

! !

A)

B)

C)

! AA!

Figu

re 7

.2: C

hang

es in

HA

DS

and

Dem

oral

izat

ion

scal

es w

ith IM

ket

amin

e 1

mg/

kg.

! A=!

Figure 7.3: Pearson’s correlation between change in depression and pain

scores.

Correlation was based in the first 6 injections, which were not administered in a weekly basis, but according to

recurrence of depressive symptoms.

!!!!!!!!!!!!!!!W(22%-&.8('!*(%))8*8%'.!9V%&2,('X,!

B>!

!!!!!!!!!!!.86%!9+>!!

! AE!

The improvement in the patient’s mood was noticed by her family, even on the phone when

talking to her sister. Also, her daughter noted she was calmer and more patient. In visits to

A.’s house to collect mood assessments these reported comments were verified.

After a total of 7 injections, A. asked for dosing before recurrence of her depressive

symptoms. As her depression was in remission for 7 days, with MADRS starting to increase 8

to 10 days post-injection, the patient started to receive ketamine on a weekly basis. With the

treatment at every 7 days, following the same protocol for the previous injections, she

obtained remission of MDD. There were two occasions, when her MADRS scores increased

to > 10, when she was unable to attend the clinic appointments, with delays between dosings

of 8 and 9 days respectively. Using this method of administration, her depression has

continued to respond to weekly ketamine dosing over 8 months (Figure 7.4). During this time,

the patient had changes in her cancer treatment, without demonstrating changes in her

response to ketamine injections for her depression. Except for the two time-points

abovementioned, the patient has maintained remission, with no complaints of cognitive

impairment. Also, side effects (such as depersonalization, inability to move, and dizziness)

were less notable with this dosage regimen.

! =:!

Figu

re 7

.4: A

ntid

epre

ssan

t res

pons

e to

repe

ated

dos

ing,

at i

nter

vals

of 7

day

s and

> 7

day

s.

!

D

osin

g at

8 –

10

day

inte

rval

s

Dos

ing

at 7

-day

inte

rval

s

! =<!

7.3 Discussion

This is the second case report with ketamine being used as a fast acting antidepressant in

patients with a diagnosis of cancer (Stefanczyk-Sapieha et al., 2008). The present report

extends information on ketamine use by assessing simultaneous effects on mood and pain

ratings, and by demonstrating the reproducibility of the antidepressant response with multiple

repeated doses.

The patient had remission (MADRS < 10; Hawley et al., 2002) of her depressive symptoms

within one hour after receiving the first injection of IM ketamine at a dose of 1mg/kg. This

finding is consistent with previous research, which noted a rapid mood response for more than

70% of patients within 24 hours (aan het Rot et al., 2010; Zarate et al., 2006). A.’s

improvement was sustained for 6 to 7 days, with depression scores gradually returning to

baseline, which was also consistent with recent studies (aan het Rot et al., 2010; Berman et

al., 2000). Some authors have reported the fact that some patients present a sustained mood

response for even longer periods of time (aan het Rot et al., 2010; Messer and Haller, 2010).

Most previous researchers used ketamine intravenously (usually over 40 minutes) rather than

IM injections, as used in this case. In a recent study, pharmacokinetic simulations suggested

comparable exposures after IV infusion and IM injection (Glue et al., 2011). In the present

case, the dose used (1mg/kg) was higher than those administered in most published cases,

which used 0.5mg/kg of Ketamine (aan het Rot et al., 2010; Zarate et al., 2006; Berman et al.,

2000), and the optimal dose for MDD treatment is still unclear. Changes in the

Demoralization Scale and HADS ratings were consistent with the MADRS data.

The analgesic effects of ketamine were also fast in onset, however its durability was variable

after 24 hours post-injection. Changes in pain scores and depression ratings were most closely

correlated 1 hour post-dosing (Pearson’s r = 0.61) with progressively weaker correlations over

the next 168 hours. This finding suggests that the sustained antidepressant effects of ketamine

are mediated differently from the mechanisms associated with immediate changes in pain and

mood. One possible mechanism may be via stimulation of mTOR signaling and via synaptic

protein synthesis (Duman et al., 2012).

A. did not experience tachyphylaxis to repeated ketamine injections, as demonstrated by the

consistent improvement in her mood scores at every injection and by her sustained remission

with weekly dosages. Messer et al., 2010 reported similar findings, with maintenance of

remission in one patient for more than 15 months, with ketamine infusions given every 3

weeks. In contrast, it should be noted that Liebrenz et al., 2009 reported a different response

! =?!

to a second ketamine infusion, with the same route of administration and dose for both, in one

patient. The subject in their study had worsening of depressive symptoms 2 days after the

second infusion, while after the first dosage the patient had presented a response sustained for

7 days. This data suggests that individual differences may influence mood responses to

repeated doses of ketamine.

According to Phelps et al., 2009 the patient’s positive mood response to ketamine might be

related to her family history of alcoholism, affecting 2 of her second-degree relatives. In the

abovementioned study, ketamine response was compared between 2 groups of depressed

subjects, one with family histories of alcoholism and the second without. Their results suggest

that a family history of alcohol dependence may be a marker for mood response to an NMDA

antagonist but, as this was the first study analyzing family history of alcohol dependence in

patients with MDD, it requires confirmation.

Administration by IM injection may have benefits when compared to IV infusion, such as:

• faster administration,

• less discomfort to patients,

• reduced costs, without the need of a pump for the infusion.

These aspects, together with her positive outcome, support the use of IM injections in future

research.

7.4 Conclusions

The use of a 1 mg/kg IM dose was well tolerated and achieved remission of depressive

symptoms. The optimal dose for the treatment of MDD has not yet been established (Glue et

al., 2011), and further data are required to define this. Considering that standard oral

antidepressants have a delay in their response of usually 4 to 6 weeks, these findings may

justify the use of IM ketamine as an antidepressant agent for a population with a short life

expectancy, as described in this case report. Also, this case illustrates the potential for

repeated use of ketamine, with maintenance of response and no complications during the

treatment. Future research is also needed to clarify the role of ketamine as a long-term

treatment option.

!

!

!

! =D!

CHAPTER 8!

!

!

Conclusions!!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

! =@!

8.1 Conclusions!

Ketamine, administered intramuscularly in a dose of 1mg/kg was effective as an

antidepressant agent. In the patient presented in this thesis, there was an improvement in

mood within 1 hour post-injection, which was maintained for up to one week, consistent with

results from published data from RCTs and case reports/ case series (Chapter 5).

!

Although most studies used intravenous administration of ketamine, intramuscular injections

have also been used successfully. This route of administration was chosen due to its simpler

and more convenient administration, with no need of an infusion pump. This choice made its

administration easy and well accepted by the patient. Side effects, such as depersonalization,

dizziness, and elevation in blood pressure, were reported, but none of them required treatment

in the previous studies reviewed in this thesis, nor in the case report presented here. Also, side

effects disappeared within two hours post-administration. This indicates that ketamine

administration should be done in a hospital, but patients might be safely discharged after side

effects are gone and vital signs are stable. The patient’s antidepressant response was

consistent after repeated injections, with no evidence of tolerance. Although this may be an

inconvenient treatment method for long term use, the observation that tolerance was not

developed means that long-term use can be considered if no other treatments are effective or

tolerated. The positive outcome with injections on a weekly basis, with maintenance of

remission of symptoms (which means the patient did not meet criteria for MDD with

continuous injections) is very promising.!

!

Although other studies must be done in this population, the promising results for ketamine has

indicate that it can be a useful medication for terminal cancer patients with depression. One

unexpected finding from this thesis was the very limited number of high quality treatment

trials from depression in cancer patients (Chapter 3). Potentially, the availability of a new

drug treatment may stimulate research in this area. For patients with terminal cancer, time is

probably the most pressing factor when they have depressive episodes, as current

antidepressant treatments take a relatively long time for therapeutic effect to occur. Then, a

fast acting antidepressant would be important to improve quality of life in terminal cancer

individuals with depression, and provide their family more positive memories after their loved

ones are gone. !

!

!

!

! ="!

CHAPTER 9

References

! =#!

9.1 References

1- aan het Rot M, Collins KA, Murrough JW, Perez AM, Reich DL, Charney DS,

Mathew SJ: Safety and efficacy of repeated-dose intravenous ketamine for treatment-resistant

depression. Biol Psychiatry 2010;67:139-145.

2- Abbass A, Driessen E: The efficacy of short-term psychodynamic psychotherapy for

depression: a summary of recent findings. Acta Psychiatr Scand 2010;121:398-399.

3- Abbott NJ, Rönnbäck L, Hansson E: Astrcyte-endothelial interactions at the blood-

brain barrier. 2006;7:41-53.

4- Akechi T, Okuyama T, Onishi J, Morita T, Furukawa TA: Psychotherapy for

depression among incurable cancer patients. Cochrane Database Syst Rev 2008 [in press].

5- Akechi T, Okuyama T, Sugawara Y, Nakano T, Shima Y, Uchitomi Y: Major

depression, adjustment disorders, and post-traumatic stress disorder in terminally ill cancer

patients: associated and predictive factors. J Clin Oncol 2004;22:1957-1965.

6- American Psychiatric Association (1980). Diagnostic and statistical manual of mental

disorders (3rd ed). Washington, DC: Author.

7- American Psychiatric Association (1987). Diagnostic and statistical manual of mental

disorders (3rd ed., Revised). Washington, DC: Author.

8- American Psychiatric Association (2000). Diagnostic and statistical manual of mental

disorders (4th ed., Text Revision). Washington, DC: Author.

9- Amsterdam JD, Shults J: Efficacy and mood conversion rate of short-term fluoxetine

monotherapy of bipolar II major depressive episode. J Clin Psychopharmacol 2010;30:306-

311.

10- Amsterdam JD, Shults J: MAOI efficacy and safety in advanced stage treatment-

resistant depression: a retrospective study. J Affect Disord 2005;89:183-188.

11- Anderson IM, Parry-Billings M, Newsholme EA, Poortmans JR, Cowen PJ:

Decreased plasma tryptophan concentration in major depression: relationship to melancholia

and weight loss. J Affect Disord 1990;20:185-191.

12- Anderson JL: The immune system and major depression. Adv Neuroimmunol

1996;6:119-129.

13- Ayuso-Mateos JL: Global burden of bipolar disorder in the year 2000. Global Program

on Evidence for Health Policy. World Health Organization 2006. In

www.who.int/healthinfo/statistics/bod_bipolar.pdf Accessed on 03 January 2012.

14- Baeken C, De Raedt R: Neurobiological mechanisms of repetitive transcranial

magnetic stimulation on the underlying neurocircuitry in unipolar depression. Dialogues Clin

Neurosci 2011;13:139-145.

! =A!

15- Bailine S, Fink M, Knapp R, Petrides G, Hussain MM, Rasmussen K, Sampson S,

Mueller M, McClintock SM, Tobias KG, Kellner CH: Electroconvulsive therapy is equally

effective in unipolar and bipolar depression. Acta Psychiatr Scand 2010;121:431-436.

16- Bair MJ, Robinson RL, Katon W, Kroenke K: Depression and pain comorbidity: a

literature review. Arch Intern Med 2003;163:2433-2445.

17- Bar-Sela G, Atid L, Danos S, Gabay N, Epelbaum R: Art therapy improved depression

and influenced fatigue levels in cancer patients on chemotherapy. Psychooncology

2007;16:980-984.

18- Bell E, Mathieu G, Racine E: Preparing the ethical future of deep brain stimulation.

Surg Neurol 2009;72:577-786.

19- Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, Krystal

JH: Antidepressant effects of ketamine in depressed patients. Biol Psychiatry 2000;47:351-

354.

20- Berrettini WH, Nurnberger Jr JI, Hare TA, Simmons-Alling S, Gershon ES, Post RM:

Reduced plasma and CSF gamma-aminobutyric acid in affective illness: effect of lithium

carbonate. Biol Psychiatry 1983;18:185-194.

21- Berridge CW, Waterhouse BD: The locus coeruleus-noradrenergic system:

modulation of behavioral state and state-dependent cognitive process. Brain Res Rev

2003;42:33-84.

22- Bijl RV, Ravelli A: Psychiatric morbidity, service use, and need for care in the general

population: results of The Netherlands Mental Health Survey and Incidence Study. Am J

Public Health 2000;90:602-607.

23- Bjelland I, Dahl AA, Haug TT, Neckelmann D: The validity of the Hospital Anxiety

and Depression Scale. An updated literature review. J Psychosom Res 2002;52:69-77.

24- Black DW, Winokur G, Nasrallah A: Mortality in patients with primary unipolar

depression, secondary unipolar depression, and bipolar affective disorder: a comparison with

general population mortality. Int J Psychiatry Med 1987;17:351-360.

25- Blomstedt P, Sjöberg RL, Hansson M, Bodlund O, Hariz MI: Deep brain stimulation

in the treatment of depression. Acta Psychiatr Scand 2011;123:4-11.

26- Bonanno FG: Ketamine in war/ tropical surgery (a final tribute to the racemic

mixture). Injury 2002;33:323-327.

27- Bos J: The rise and decline of character: humoral psychology in ancient and early

modern medical theory. Hist Human Sci 2009;22:1-27.

! ==!

28- Bosmans JE, de Bruijne MC, de Boer MR, van Hout H, van Steenwijk P, van Tulder

MW: Health care costs of depression in primary care patients in The Netherlands. Fam Pract

2010;27:542-548.

29- Boulenger JP, Hermes A, Huusom AK, Weiller E: Baseline anxiety effect on outcome

of SSRI treatment in patients with severe depression: escitalopram vs paroxetine. Curr Med

Res Opin 2010;26:605-614.

30- Boyes AW, Girgis A, D'Este C, Zucca AC: Flourishing or floundering? Prevalence

and correlates of anxiety and depression among a population-based sample of adult cancer

survivors 6months after diagnosis. J Affect Disord 2011;135:184-192.

31- Brambilla P, Perez J, Barale F, Schettini G, Soares JC: GABAergic dysfunction in

mood disorders. Mol Psychiatry 2003;8:721-737.

32- Breen SJ, Baravelli CM, Schofield PE, Jefford M, Yates PM, Aranda SK: Is symptom

burden a predictor of anxiety and depression in patients with cancer about to commence

chemotherapy? Med J Aust. 2009;190:99-104.

33- Britt GC, McCance-Katz EF: A brief overview of the clinical pharmacology of “club

drugs”. Subst Use Misuse 2005;40:1189-1201.

34- Bruera E, Carraro S, Roca E, Barugel M, Chacon R: Double-blind evaluation of the

effects of mazindol on pain, depression, anxiety, appetite, and activity in terminal cancer

patients. Cancer Treat Rep 1986;70:295-298.

35- Byers AL, Yaffe K: Depression and risk of developing dementia. Nat Rev Neurol

2011;7:323-331.

36- Caric D, Kaithackachalil S, Kim J, Nickell PV, Nathan S: A case study of a patient

with a medication-resistant major depression treated with ketamine infusion followed by

pharmacotherapy alone vs combined ECT and pharmacotherapy. J ECT 2007;23:51.

37- Castelli L, Binaschi L, Caldera P, Mussa A, Torta R: Fast screening of depression in

cancer patients: the effectiveness of the HADS. Eur J Cancer Care (Engl) 2011;20:528-533.

38- Chakrabarti I, Giri A, Majumdar K, DE A: An unusual case of thyroid papillary

carcinoma with solitary cerebral metastasis presenting with neurological symptoms. Turk

Patoloji Derg 2011;27:154-156.

39- Chouinard G, Saxena BM, Nair NP, Kutcher SP, Bakish D, Bradwejn J, Kennedy SH,

Sharma V, Remick RA, Kukha-Mohamad SA, et al. A Canadian multicentre placebo-

controlled study of a fixed dose of brofaromine, a reversible selective MAO-A inhibitor, in

the treatment of major depression. J Affect Disord 1994;32:105-114.

40- Coentre R, Barrocas D, Chendo I, Abreu M, Lvy P, Maltez J, Figueira ML:

Electroconvulsive therapy: myths and evidences. Acta Med Port 2009;22:275-280.

! =E!

41- Colleoni M, Mandala M, Peruzzotti G, Robertson C, Bredart A, Goldhirsch A:

Depression and degree of acceptance of adjuvant cytotoxic drugs. Lancet 2000;356:1326-

1327.

42- Coppen AJ: The biochemistry of affective disorders. Br J Psychiatry 1967;113:1237-

1264.

43- Costa D, Mogos I, Toma T: Efficacy and safety of mianserin in the treatment of

depression of women with cancer. Acta Psychiatr Scand Suppl 1985;320:85-92.

44- Cuijpers P, Andersson G, Donker T, van Straten A: Psychological treatment of

depression: results of a series of meta-analyses. Nord J Psychiatry 2011;65:354-3.64.

45- Dadu S, Mishra LS, Agrasal M, Chandola HC: Comparative clinical study of effect of

neostigmine and ketamine for postoperative analgesia. J Indian Med Assoc 2011;109:308-

311.

46- Dagyt" G, Den Boer JA, Trentani A: The cholinergic system and depression. Behav

Brain Res 2011;221:574-582.

47- Daly RW: Before depression: the medieval vice of acedia. Psychiatry 2007;70:30-51.

48- Davison K: Historical aspects of mood disorders. Psychiatry 2009;8:47-51.

49- Dell'osso B, Camuri G, Castellano F, Vecchi V, Benedetti M, Bortolussi S, Altamura

AC: Meta-Review of Metanalytic Studies with Repetitive Transcranial Magnetic Stimulation

(rTMS) for the Treatment of Major Depression. Clin Pract Epidemiol Ment Health

2011;7:167-177.

50- Deng GF, Zheng JP, Wang S, Tian B, Zhang SG: Remifentanil combined with low-

dose ketamine for postoperative analgesia of lower limb fracture: a double-blind, controlled

study. Chin J Traumatol 2009;12:223-227.

51- Denk MC, Rewerts C, Holsboer F, Erhardt-Lehmann A, Turck CW: Monitoring

ketamine treatment response in a depressed patient via peripheral mammalian target of

rapamycin activation. Am J Psychiatry 2011;168:751-752.

52- Diazgranados N, Ibrahim L, Britsche NE, Newberg A, Kronstein P, Khalife S,

Kammerer WA, Quezado Z, Luckenbaugh DA, Salvadore G, Machado-Vieira R, Manji HK,

Zarate CA Jr: A randomized add-on trial of an N-methyl-D-aspartate antagonist in treatment-

resistant bipolar depression. Arch Gen Psychiatry 2010;67:793-802.

53- Dillon P, Copeland J, JansenK: Patterns of use and harms associated with non-medical

ketamine use. Drug Alcohol Depend 2003;69:23-28.

54- Dorr AE, Debonnel G: Effect of vagus nerve stimulation on serotonergic and

noradrenergic transmission. J Pharmacol Exp Ther 2006;318:890-898.

! E:!

55- Duman RS, Li N, Liu RJ, Duric V, Aghajanian G: Signaling pathways underlying the

rapid antidepressant actions of ketamine. Neuropharmacology 2012;62:35-41.

56- Dunlop BW, Nemeroff CB: The role of dopamine in the pathophysiology of

depression. Arch Gen Psychiatry 2007;64:327-337.

57- Dwight-Johnson M, Ell K, Lee P: Can collaborative care address the needs of low-

income latinas with comorbid depression and cancer? Results from a randomized pilot study.

Psychosomatics 2005;46:224-232.

58- Ell K, Xie B, Kapetanovic S, Quinn DI, Lee P, Wells A, Chou C: One-year follow-up

of collaborative depression care for low-income, predominantly Hispanic patients with

cancer. Psychiatr Serv 2011;62:162-170.

59- Ell K, Xie B, Quon B, Quinn DI, Dwight-Johnson M, Lee P: Randomized controlled

trial of collaborative care management of depression among low-income patients with cancer.

J Clin Oncol 2008;26:4488-4496.

60- Evans RL, Connis RT: Comparison of brief group therapies for depressed cancer

patients receiving radiation treatment. Public Health Rep 1995;110:306-311.

61- Feng CY, Chu H, Chen CH, Chang YS, Chen TH, Chou YH, Chang YC, Chou KR:

The Effect of Cognitive Behavioral Group Therapy for Depression: A Meta-Analysis 2000-

2010. Worldviews Evid Based Nurs 2012;9:2-17.

62- Fiedorowicz JG, Palagummi NM, Forman-Hoffman VL, Miller DD, Haynes WG:

Elevated prevalence of obesity metabolic syndrome, and cardiovascular risk factors in bipolar

disorder. Ann Clin Psychiatry 2008;20:131-137.

63- Fitzgerald PB, Daskalakis ZJ: A practical guide to the use of repetitive transcranial

magnetic stimulation in the treatment of depression. Brain Stimul 2011 [in press].

64- Gershon AA, Dannon PN, Grunhaus L: Transcranial magnetic stimulation in the

treatment of depression. Am J Psychiatry 2003;160:835-45.

65- Ghaemi SN: Treatment of rapid-cycling bipolar disorder: are antidepressants mood

destabilizers? Am J Psychiatry 2008;165:300-302.

66- Glue P, Gulati A, Le Nedelec M, Duffull S: Dose and exposure response to Ketamine

in depression. Biol Psychiatry 2011;70:e9-10.

67- Goekoop JG, de Winter RF, Wolterbeek R, van Kempen GM, Wiegant VM: Evidence

of vasopressinergic-noradrenergic mechanisms in depression with above-normal plasma

vasopressin concentration with and without psychotic features. J Psychopharmacol

2011;25:345-352.

68- Goforth HW, Holsinger T: Rapid relief of severe major depressive disorder by the use

of preoperative Ketamine and electroconvulsive therapy. J ECT 2007;23:23-25.

! E<!

69- Goodnick PJ: Bipolar depression: a review of randomized clinical trials. Expert Opin

Pharmacother 2007;8:13-21.

70- Greenberg PE, Stiglin LE, Finkelstein SN, Berndt ER: The economic burden of

depression in 1990. J Clin Psychiatry 1993;54:405-418.

71- Guidi J, Fava GA, Picardi A, Porcelli P, Bellomo A, Grandi S, Grassi L, Pasquini P,

Quartesan R, Rafanelli C, Rigatelli M, Sonino N: Subtyping depression in the medically ill by

cluster analysis. J Affect Disord 2011;132:383-388.

72- Hamilton M: Development of a rating scale for primary depressive illness. Br J Soc

Clin Psychol 1967;6:278-296.

73- Hamani C, Nóbrega JN: Deep brain stimulation in clinical trials and animal models of

depression. Eur J Neurosci 2010;32:1109-1117.

74- Harich, J. The comparative pain scale. In: http://www.tipna.org/info/documents/

ComparativePainScale.htm. Accessed on 10/03/2011.

75- Harro J, Oreland L: Depression as a spreading adjustment disorder of monoaminergic

neurons: a case for primary implication of the locus coeruleus. Brain Res Brain Res Rev

2001;38:79-128.

76- Härter M, Klesse C, Bermejo I, Schneider F, Berger M: Unipolar depression:

diagnostic and therapeutic recommendations from the current S3/National Clinical Practice

Guideline. Dtsch Arztebl Int 2010;107:700-708.

77- Hashimoto K: Emerging role of glutamate in the pathophysiology on major depressive

disorder. Brain Res Rev 2009; 61: 105-123.

78- Hasler G, van der Veen JW, Tumonis T, Meyers N, Shen J, Drevets WC: Reduced

prefrontal glutamate/glutamine and gamma-aminobutyric acid levels in major depression

determined using proton magnetic resonance spectroscopy. Arch Gen Psychiatry

2007;64:193-200.

79- Hawley CJ, Gale TM, Sivakumaran T: Defining remission by cut off score on the

MADRS: selecting the optimal value. J Affect Disord 2002;72:177-184.

80- Holland JC, Romano SJ, Heiligenstein JH, Tepner RG, Wilson MG: A controlled trial

of fluoxetine and desipramine in depressed women with advanced cancer. Psychooncology

1998;7:291-300.

81- Holland PR, Akerman S, Goadsby PJ: Cortical spreading depression-associated

cerebral blood flow changes induced by mechanical stimulation are modulated by AMPA and

GABA receptors. Cephalalgia 2010;30:519-527.

82- Holtzheimer PE 3rd, Mayberg HS: Deep brain stimulation for treatment-resistant

depression. Am J Psychiatry 2010;167:1437-1444.

! E?!

83- Huisman M: King Saul, work-related stress and depression. J Epidemiol Community

Health 2007;61:890.

84- Ibrahim L, Diazgranados N, Luckenbaugh DA, Machado-Vieira R, Baumann J,

Mallinger AG, Zarate CA Jr: Rapid decrease in depressive symptoms with an N-methyl-d-

aspartate antagonist in ECT-resistant major depression. Arch Gen Psychiatry 2010;67:793-

802.

85- Ihmsen H, Geisslinger G, Schuttler J: Stereoselective pharmacokinetics of ketamine:

R(-)-ketamine inhibits the elimination of S(+)-ketamine. Clin Pharmacol Ther 2001;70:431-

438.

86- Irwin SA, Iglewicz A: Oral ketamine for the rapid treatment of depression and anxiety

in patients receiving hospice care. J Palliat Med 2010;13:903-908.

87- Ishak WW, Ha K, Kapitanski N, Bagot K, Fathy H, Swanson B, Vilhauer J, Balayan

K, Bolotaulo NI, Rapaport MH: The impact of psychotherapy, pharmacotherapy, and their

combination on quality of life in depression. Harv Rev Psychiatry 2011;19:277-289.

88- Itoi K, Sugimoto N: The brainstem noradrenergic systems in stress, anxiety and

depression. J Neuroendocrinol 2010;22:355-361.

89- Jakobsen JC: Review: interpersonal psychotherapy is effective as an acute or

maintenance treatment for unipolar depression. Evid Based Ment Health 2011;14:108.

90- Joels M, Baram TZ: The neuro-symphony of stress. Nat Rev Neurosci 2009;10:459-

466.

91- Kahl KG, Greggersen W, Schweiger U, Cordes J, Balijepalli C, Lösch C, Moebus S:

Prevalence of the metabolic syndrome in unipolar major depression. Eur Arch Psychiatry Clin

Neurosci 2011 [in press].

92- Keddie AM: Associations between severe obesity and depression: results from the

National Health and Nutrition Examination Survey, 2005-2006. Prev Chronic Dis 2011;8:57.

93- Keller MB, Lavori PW, Mueller TI, Endicott J, Coryell W, Hirschfeld RM, Shea T:

Time to recovery, chronicity, and levels of psychopathology in major depression. A 5-year

prospective follow-up of 431 subjects. Arch Gen Psychiatry 1992;49:809-816.

94- Keller PA, McCluskey A, Morgan J, O’connor SM: The role of the HPA axis in

psychiatric disorders and CRF antagonists as potential treatments. Arch Pharm (Weinheim)

2006;339:346-355.

95- Kessing L, Andersen P: Does the risk of developing dementia increase with the

number of episodes in patients with depressive disorder and in patients with bipolar disorder?

J Neurol Neurosurg Psychiatry 2004;75:1662-1666.

! ED!

96- Kessler RC, Chiu WT, Demler O, Walters EE: Prevalence, severity and co-morbidity

of twelve-month DSM-IV disorders in the National Comorbidity Survey Replication (NCS-

R). Arch Gen Psychiatry 2005;62:617-627.

97- Ketalar- Medsafe. (date of preparation: 19 July 2010). In

www.medsafe.govt.nz/profs/datasheet/k/ketalarinj.pdf. Assessed on 11 November 2011.

98- Khawam EA, Laurencic G, Malone DA Jr: Side effects of antidepressants: an

overview. Cleve Clin J Med 2006;73:351-353, 356-361.

99- Kissane DW, Wein S, Love A, Lee XQ, Kee PL, Clarke DM: The Demoralization

Scale: a report of its development and preliminary validation. J Palliat Care 2004;20:269-276.

100- Klemm R, Hardie T: Depression in Internet and face-to-face cancer support groups: a

pilot study. Oncol Nurs Forum 2002;29:45-51.

101- Ko GJ, Kim MG, Yu YM, Jo SK, Cho WY, Kim HK: Association between depression

symptoms with inflammation and cardiovascular risk factors in patients undergoing peritoneal

dialysis. Nephron Clin Pract 2010;116:29-35.

102- Koike H, Lijima M, Chaki S: Involvement of the mammalian target of rapamycin

signaling in the antidepressant-like effect of group II metabotropic glutamate receptor

antagonists. 2011;61:1419-1423.

103- Kornstein SG, Li D, Mao Y, Larsson S, Andersen HF, Papakostas GI: Escitalopram

versus SNRI antidepressants in the acute treatment of major depressive disorder: integrative

analysis of four double-blind, randomized clinical trials. CNS Spectr 2009;14:326-333.

104- Kurz K, Schroecksnadel S, Weiss G, Fuchs D: Association between increased

tryptophan degradation and depression in cancer patients. Curr Opin Clin Nutr Metab Care

2011;14:49-56.

105- Kwok RFK, Lin J, Chan MTV, Gin T, Chiu WKY: Preoperative ketamine improves

postoperative analgesia after gynecologic laparoscopic surgery. Anesth Analg 2004;98:1044-

1049.

106- Larkin GL, Beautrais AL: A preliminary naturalistic study of low dose ketamine for

depression and suicide ideation in the emergency department. Int J Neuropsychopharmacol

2011;14:1127-1131.

107- Laskowski K, Stirling A, Mckay WP, Lim HJ: A systematic review of intravenous

ketamine for postoperative analgesia. Can J Anaesth 2011;58:911-923.

108- Lavori PW, Keller MB, Mueller TI, Scheftner W, Fawcett J, Coryell W: Recurrence

after recovery in unipolar MDD: an observational follow-up study of clinical predictors and

somatic treatment as a mediating factor. Int J Methods Psychiatr Res 1994;4:211-229.

! E@!

109- Lee TS: Transient and spontaneously-remitting complex hallucinations in a patient

with melanoma and brain metastases. 2010;51:267-270.

110- Leonard B, Maes M: Mechanistic explanations how cell-mediated immune activation,

inflammation and oxidative and nitrosative stress pathways and their sequels and

concomitants play a role in the pathophysiology of unipolar depression. Neurosci Biobehav

Rev 2012;36:764-785.

111- Lester L, Braude DA, Niles C, Crandall CS: Low-dose ketamine for analgesia in the

ED: a retrospective case series. Am J Emerg Med 2010;28:820-827.

112- Liebrenz M, Stohler R, Borgeat A: Repeated intravenous ketamine therapy in a patient

with treatment-resistant major depression. World J Biol Psychiatry 2009;10:640-643.

113- Lisanby SH, Maddox JH, Prudic J, Devanand DP, Sackeim HA: The effects of

electroconvulsive therapy on memory of autobiographical and public events. Arch Gen

Psychiatry 2000;57:581-590.

114- Loo CK, McFarquhar TF, Mitchell PB: A review of the safety of repetitive

transcranial magnetic stimulation as a clinical treatment for depression. Int J

Neuropsychopharmacol 2008;11:131-147.

115- Machado-Vieira R, Yuan P, Brutsche N, Diazgranados N, Luckenbaugh D, Manji HK,

Zarate CA Jr: Brain-derived neurotrophic factor and initial antidepressant response to an N-

methyl-D-aspartate antagonist. J Clin Psychiatry 2009;70:1662-1666.

116- Mak SK, Chan MT, Bower WF, Yip SK, Hou SS, Wu BB, Man CY: Lower urinary

tract changes in young adults using ketamine. J Urol 2011;186:610-614.

117- Malone DA Jr: Use of deep brain stimulation in treatment-resistant depression. Cleve

Clin J Med 2010;77:77-80.

118- Markowitz JC, Weissman MM: Interpersonal Psychotherapy: Past, Present and

Future. Clin Psychol Psychother 2012 Feb [in press].

119- Martin JL, Martin-Sanchez E: Systematic review and meta-analysis of vagus nerve

stimulation in the treatment of depression: Variable results based on study designs. Eur

Psychiatry 2012;27:147-155.

120- Mason K, Cottrell AM, Corrigan AG, Gillatt DA, Mitchelmore AE: Ketamine-

associated lower urinary tract destruction: a new radiological challenge. Clin Radiol

2010;65:795-800.

121- Mathers CD, Loncar D: Updated projections of global mortality and burden of disease,

2001- 2030: data sources, methods and results. Evidence and information for policy. World

Health Organization 2005. In

! E"!

http://www.who.int/healthinfo/statistics/bodprojectionspaper.pdf. Accessed on 03 January

2012.

122- Mathew SJ, Murrough JW, aan het Rot M, Collins KA, Reich DL, Charney DS:

Riluzole for relapse prevention following intravenous ketamine in treatment-resistant

depression: a pilot randomized, placebo-controlled continuation trial. Int J

Neuropsychopharmacol 2010 ;13:71-82.

123- Mayberg HS, Liotti M, Brannan SK, McGinnis S, Mahurin RK, Jerabek PA, Silva JA,

Tekell JL, Martin CC, Lancaster JL, Fox PT: Reciprocal limbic-cortical function and negative

mood: converging PET findings in depression and normal sadness. Am J Psychiatry

1999;156:675-682.

124- Mayberg HS, Lozano AM, Voon V, McNeely HE, Seminowicz D, Hamani C,

Schwalb JM, Kennedy SH: Deep brain stimulation for treatment-resistant depression. Neuron

2005;45:651-660.

125- McEwen BS: Stress and hippocampal plasticity. Annu Rev Neurosci 1999;22:105-

122.

126- McEwen BS, Magarinos AM: Stress and hippocampal plasticity: implications for the

pathophysiology of affective disorders. Hum Psychopharmacol 2001;16:7-19.

127- Mercadante S, Porzio G, Gebbia V: Spinal analgesia for advanced cancer patients: An

update. Crit Rev Oncol Hematol 2011;16:in press.

128- Messer MM, Haller IV: Maintenance ketamine treatment produces long-term recovery

from depression. Primary Psychiatry 2010;17:48-50.

129- Messer M, Haller IV, Larson P, Pattison-Crisostomo J, Gessert CE: The use of a series

of ketamine infusions in two patients with treatment-resistant depression. J Neuropsychiatry

Clin Neurosci 2010;22:442-444.

130- Middlemiss DN, Price GW, Watson JM: Serotonergic targets in depression. Curr Opin

Pharmachol 2002;2:18-22.

131- Miller AH, Raison CL: Immune system contributions to the pathophysiology of

depression. FOCUS 2008;6:36-45.

132- Minichino A, Bersani FS, Capra E, Pannese R, Bonanno C, Salviati M, Delle Chiaie

R, Biondi M: ECT, rTMS, and deepTMS in pharmacoresistant drug-free patients with

unipolar depression: a comparative review. Neuropsychiatr Dis Treat 2012;8:55-64.

133- Mitchell AJ: New developments in the detection and treatment of depression in cancer

settings. Prog Neurol Psychiatry 2011;10: in press.

134- Mitchell AJ, Chan M, Bhatti H, Halton M, Grassi L, Johansen C, Meader N:

Prevalence of depression, anxiety, and adjustment disorder in oncological, haematological,

! E#!

and palliative-care settings: a meta-analysis of 94 interview-based studies. Lancet Oncol

2011;12:160-174.

135- Mitchell PB, Malhi GS: Bipolar depression: phenomenological overview and clinical

characteristics. Bipolar Disord 2004;6:530-539.

136- Mohler H: The GABA system in anxiety and depression and its therapeutic potential.

Neuropharmacology 2012;62:42-53.

137- Mongini F, Ciccone G, Ceccarelli M, Baldi I, Ferrero L: Muscle tenderness in

different types of facial pain and its relation to anxiety and depression: A cross-sectional

study on 649 patients. Pain 2007;131:106-111.

138- Montgomery SA, Asberg M: A new depression scale designed to be sensitive to

change. Br J Psychiatry 1979;134:382-389.

139- Montgomery A, Briley M: Noradrenergic symptom cluster in depression.

Neuropsychiatr Dis Treat 2011;7:1-2.

140- Murray CJL, Lopez AD: The global burden of disease: a comprehensive assessment of

mortality and disability from diseases, injuries, and risk factors in 1990 and projected to 2020.

Harvard School of Public Health 1996.

141- Nikish G, Mathe AA: CSF monoamine metabolites and neuropeptides in depressed

patients before and after electroconvulsive therapy. Eur Psychiatry 2008;23:356-359.

142- Noppers I, Niesters M, Swartjes M, Bauer M, Aarts L, Geleijnse N, Mooren R, Dahan

A, Sarton E: Absence of long-term analgesic effect from a short-term S-ketamine infusion on

fibromyalgia pain: a randomized, prospective, double blind, active placebo-controlled trial.

Eur J Pain 2011;15:942-949.

143- Nutt DJ: The role of dopamine and norepinephrine in depression and antidepressant

treatment. J Clin Psychiatry 2006;67:3-8.

144- Nutt D, Demyttenaere K, Janka Z, Aarre T, Bourin M, Canonico PL, Carrasco JL,

Stahl S: The other face of depression, reduced positive affect: the role of catecholamines in

causation and cure. J Psychopharmacol 2007;21:461-471.

145- O’Connor MF, Irwin MR, Seldon J, Kwan L, Ganz PA: Pro-inflammatory cytokines

and depression in a familial cancer registry. Psychooncology 2007;16:499-501.

146- Ohayon MM: Pain sensitivity, depression, and sleep deprivation: Links with

serotoninergic disfunction. J Psychiatr Res 2009;43:1243-1245.

147- Oken MM, Creech RH, Tormey DC, Horton J, Davis TE, McFadden ET, Carbone PP:

Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol

1982;5:649-655.

! EA!

148- Osby U, Brandt L, Correia N, Ekbom A, Sparén P: Excess mortality in bipolar and

unipolar disorder in Sweden. Arch Gen Psychiatry 2001;58:844-850.

149- Pacchiarotti I, Mazzarini L, Colom F, Sanchez-Moreno J, Girardi P, Kotzalidis GD,

Vieta E: Treatment-resistant bipolar depression: towards a new definition. Acta Psychiatr

Scand 2009;120:429-440.

150- Pacchiarotti I, Valentí M, Bonnin CM, Rosa AR, Murru A, Kotzalidis GD, Nivoli

AM, Sánchez-Moreno J, Vieta E, Colom F: Factors associated with initial treatment response

with antidepressants in bipolar disorder. Eur Neuropsychopharmacol 2011;21:362-369.

151- Paslakis G, Gilles M, Meyer-Lindenberg A, Deuschle M: Oral administration of the

NMDA receptor antagonist S-ketamine as add-on therapy of depression: a case series.

Pharmacopsychiatry 2010;43:33-35.

152- Paul IA, Skolnick P: Glutamate and depression: clinical and preclinical studies. Ann

NY Acad Sci 2003;1003:250-272.

153- Paul R, Schaaff N, Padberg F, Möller HJ, Frodl T: Comparison of racemic ketamine

and S-ketamine in treatment-resistant major depression: report of two cases. World J Biol

Psychiatry 2009;10:241-244.

154- Petty F, Kramer GL, Gullion CM, Rush AJ: Low plasma gamma-aminobutyric acid

levels in male patients with depression. Biol Psychiatry 1992;32:354-363.

155- Petty F, Steinberg J, Kramer GL, Fulton M, Moeller FG: Desipramine does not alter

plasma GABA in patients with major depression. J Affect Disord 1993;29:53-56.

156- Pezzella G, Moslinger-Gehmayr R, Contu A: Treatment of depression in patients with

breast cancer: a comparison between paroxetine and amitriptyline. Breast Cancer Res Treat

2001;70:1-10.

157- Phelps LE, Brutsche N, Moral JR, Luckenbaugh DA, Manji HK, Zarate CA Jr: Family

history of alcohol dependence and initial antidepressant response to an M-methyl-D-aspartate

antagonist. Biol Psychiatry 2009;65:181-184.

158- Pincus HA, Pettit AR: The societal costs of chronic major depression. J Clin

Psychiatry 2001;62:5-9.

159- Popoli M, Yan Z, McEwen BS, Sanacora G: The stressed synapse: the impact of stress

and glucocorticoids on glutamate transmission. Nat Rev Neurosci 2011;13:22-37.

160- Price RB, Nock MK, Charney DS, Mathew SJ: Effects of intravenous ketamine on

explicit and implicit measures of suicidality in treatment-resistant depression. Biol Psychiatry

2009;66:522-526.

! E=!

161- Rayner L, Price A, Evans A, Valsraj K, Hotopf M, Higginson IJ: Antidepressants for

the treatment of depression in palliative care: systematic review and meta-analysis. Palliat

Med 2011;25:36-51.

162- RevMan 5.1. Cochrane Collaboration 2008. In: www.cc-ims.net/RevMan. Accessed

on 19 December 2011.

163- Roscoe JA, Morrow GR, Hickok JT, Mustian KM, Griggs JJ, Matteson SE, Bushunow

P, Qazi R, Smith B: Effect of paroxetine hydrochloride (Paxil ®) on fatigue and depression in

breast cancer patients receiving chemotherapy. Breast Cancer Res Treat 2005;89:243-249.

164- Rudolph RL, Feiger AD: A double-blind, randomized, placebo-controlled trial of

once-daily venlafaxine extended release (XR) and fluoxetine for the treatment of depression. J

Affect Disord 2000;56:171-181.

165- Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg AA, Stewart JW, Warden D,

Niederehe G, Thase ME, Lavori PW, Lebowitz BD, McGrath PJ: Acute and long-term

outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report.

Am J Psychiatry 2006;163:1905-1917.

166- Sackeim HA, Rush AJ, George MS, Marangell LB, Husain MM, Nahas Z, Johnson

CR, Seidman S, Giller C, Haines S, Simpson RK Jr, Goodman RR: Vagus nerve stimulation

(VNS) for treatment-resistant depression: efficacy, side effects, and predictors of outcome.

Neuropsychopharmacology 2001;25:713-728.

167- Salvadore G, van der Veen JW, Zhang Y, Marenco S, Machado-Vieira R, Baumann J,

Ibrahim LA, Luckenbaugh DA, Shen J, Drevets WC, Zarate CA: An investigation of amino-

acid neurotransmitters as potential predictors of clinical improvement to ketamine in

depression. Int J Neuropsychopharmacol 2011; 1:1-10.

168- Sanacora G, Gueorguieva R, Epperson CN, Wu YT, Appel M, Rothman DL, Krystal

JH, Mason GF: Subtype-specific alterations of gamma-aminobutyric acid and glutamate in

patients with major depression. Arch Gen Psychiatry 2004;61:705-713.

169- Sanacora G, Treccani G, Popoli M: Towards a glutamate hypothesis of depression: An

emerging frontier of neuropsychopharmacology for mood disorders. Neuropharmacology

2012;62:63-77.

170- Santiago-Rodriguez E, Sales-Carmona V, Ramos-Ramirez R: Risk factors for

therapeutic noncompliance of patients with epilepsies. Gac Med Mex 2002;138:241-246.

171- Sara SJ: The locus coeruleus and noradrenergic modulation of cognition. Nat Rev

Neurosci 2009;10:211-223.

! EE!

172- Satyanarayana S, Enns MW, Cox BJ, Sareen J: Prevalence and correlates of chronic

depression in the Canadian community health survey: mental health and well-being. Can J

Psychiatry 2009;54:389-398.

173- Savard J, Simard S, Giguere I, Ivers H, Morin CM, Maunsell E, Gagnon P, Robert J,

Marceau D: Randomized clinical trial on cognitive therapy for depression in women with

metastatic breast cancer: psychological and immunological effects. Palliat Support Care

2006;4:219-237.

174- Schiepers OJ, Wichers MC, Maes M: Citokines and major depression. Prog

Neuropsychopharmacol Biol Psychiatry 2005;29:637-638.

175- Schoeyen HK, Birkenaes AB, Vaaler AE, Auestad BH, Malt UF, Andreassen OA,

Morken G: Bipolar disorder patients have similar levels of education but lower socio-

economic status than the general population. J Affect Disord 2011;129:68-78.

176- Schofield DJ, Kelly SJ, Shrestha RN, Callander EJ, Percival R, Passey ME: How

depression and other mental health problems can affect future living standards of those out of

the labor force. Aging Ment Health 2011;15:654-662.

177- Scott KM, Browne MAO, Wells JE: Prevalence, impairment and severity of 12-month

DSM-IV major depressive episodes in Te Rau Hinengaro: New Zealand Mental Health

Survey 2003/4. Aust N Z Psychiatry 2010;44:750-758.

178- Shahani R, Streutker C, Dickson B, Stewart RJ: Ketamine-associated ulcerative

cystitis: a new clinical entity. Urology 2007;69:810-812.

179- Shopsin B: Enhancement of the antidepressant response to L-tryptophan by a liver

pyrrolase inhibitor: a rational treatment. Neuropsychobiology 1978;4:188-192.

180- Sidor MM, Macqueen GM: Antidepressants for the acute treatment of bipolar

depression: a systematic review and meta-analysis. J Clin Psychiatry 2011;72:156-167.

181- Sih K, Campbell SG, Tallon JM, Magee K, Zed PJ. Ketamine in adult emergency

medicine: controversies and recent advances. Ann Pharmacother. 2011 Dec;45(12):1525-34.

182- Skolnick P, Popik P, Trullas R: Glutamate based antidepressants: 20 years on. Trends

Pharmacol Sci 2009; 30: 563-569.

183- Sofia RD, Harakal JJ: Evaluation of ketamine HCl for anti-depressant activity. Arch

Int Pharmacodyn Ther 1975;214:68-74.

184- Souery D, Serretti A, Calati R, Oswald P, Massat I, Konstantinidis A, Linotte S,

Bollen J, Demyttenaere K, Kasper S, Lecrubier Y, Montgomery S, Zohar J, Mendlewicz J:

Switching antidepressant class does not improve response or remission in treatment-resistant

depression. J Clin Psychopharmacol 2011;31:512-516.

185- Statistics New Zealand. In: www.stats.govt.nz. Accessed on 20 January 2012.

! <::!

186- Stefanczyk-Sapieha L, Oneschuk D, Demas M: Intravenous ketamine “burst” for

refractory depression in a patient with advanced cancer. J Palliat Med 2008;11:1268-1271.

187- Strigo IA, Simmons AN, Matthews SC, Craig AD, Paulus MP: Association of major

depressive disorder with altered functional brain response during anticipation and processing

of heat pain. Arch Gen Psychiatry 2008;65:1275-1284.

188- Strong V, Waters R, Hibberd C, Murray G, Wall L, Walker J, McHugh G, Walker A,

Sharpe M: Management of depression for people with cancer (SmaRT oncology 1): a

randomized trial. Lancet 2008;372:40-48.

189- Tolvon- Medsafe. In www.medsafe.govt.nz/profs/datasheet/t/Tolvontab.pdf. Assessed

on 1 February 2012.

190- Trevino K, McClintock SM, Husain MM: A review of continuation electroconvulsive

therapy: application, safety and efficacy. J ECT 2010;26:186-195.

191- Trivedi M, Rush AJ, Wisniewski SR, Nierenberg AA, Warden D, Ritz L, Norquist G,

Howland RH, Lebowitz B, McGrath PJ, Shores-Wilson K, Biggs MM, Balasubramani GK,

Fava M: Evaluation of outcomes with citalopram for depression using measurement-based

care in STAR*D: implications for clinical practice. Am J Psych 2006;163:28-40.

192- Trivedi RB, Nieuwsma JA, Williams JW Jr: Examination of the utility of

psychotherapy for patients with treatment resistant depression: a systematic review. J Gen

Intern Med 2011;26:643-650.

193- UK ECT Review group: Efficacy and safety of electroconvulsive therapy in

depressive disorders: a systematic review and meta-analysis. Lancet 2003;361:799-808.

194- Ustün TB, Ayuso-Mateos JL, Chatterji S, Mathers C, Murray CJ: Global burden of

depressive disorders in the year 2000. Br J Psychiatry 2004;184:386-392.

195- Valentine GW, Mason GF, Gomez R, Fasula M, Watzl J, Pittman B, Krystal JH,

Sanacora G: The antidepressant effect of ketamine is not associated with changes in occipital

amino acid neurotransmitter content as measured by [(1)H]-MRS. Psychiatry Res

2011;191:122-127.

196- van Calker D, Zobel I, Dykierek P, Deimel CM, Kech S, Lieb K, Berger M, Schramm

E: Time course of response to antidepressants: predictive value of early improvement and

effect of additional psychotherapy. J Affect Disord 2009;114:243-253.

197- van Den Eede F, Claes S: Mechanisms of depression: role of the HPA axis. Drug

Discov Today Dis Mech 2004;1:413-418.

198- van Heeringen K, Zivkov M: Pharmacological treatment of depression in cancer

patients. A placebo-controlled study of mianserin. Br J Psychiatry 1996;169:440-443.

! <:<!

199- Wassermann EM: Side effects of repetitive transcranial magnetic stimulation. Depress

Anxiety 2000;12:124-129.

200- Waters SJ, Keefe FJ, Strauman TJ: Self-discrepancy in chronic low back pain: relation

to pain, depression, and psychological distress. J Pain Symptom Manage 2004 Mar;27:251-9.

201- Weilburg JB: An overview of SSRI and SNRI therapies for depression. Manag care

2004;13:25-33.

202- Weiner M, Warren L, Fiedorowicz JG: Cardiovascular morbidity and mortality in

bipolar disorder. Ann Clin Psychiatry 2011;23:40-47.

203- White PF, Schüttler J, Shafer A, Stanski DR, Horai Y, Trevor AJ: Comparative

pharmacology of the ketamine isomers. Studies in volunteers. Br J Anaesth 1985;57:197-203.

204- White VM, Macvean ML, Grogan S, D'Este C, Akkerman D, Ieropoli S, Hill DJ,

Sanson-Fisher R: Can a tailored telephone intervention delivered by volunteers reduce the

supportive care needs, anxiety and depression of people with colorectal cancer? A randomised

controlled trial. Psychooncology 2011 [in press].

205- Williams JBW: A structured interview guide for the Hamilton Depression Rating

Scale. Arch Gen Psychiatry 1988;45:742-747.

206- Williams S, Dale J: The effectiveness of treatment for depression/depressive

symptoms in adults with cancer: a systematic review. Br J Cancer 2006;94:372-390.

207- World Health Statistics 2007.

www.who.int/entity/whosis/whostat2007_10highlights.pdf. Assessed on 22 November 2011.

208- Yan XY, Huang SM, Huang CQ, Wu WH,Qin Y: Marital status and risk for late life

depression: a meta-analysis of the published literature. J Int Med Res 2011;39:1142-1154.

209- Yirmiya R, Weidenfeld J, Pollak Y, Morag M, Morag A, Avitsur R, Barak O,

Reichenberg A, Cohen E, Shavit Y, Ovadia H: Cytokines, "depression due to a general

medical condition," and antidepressant drugs. Adv Exp Med Biol 1999;461:283-316.

210- Young SN, Smith SE, Pihl RO, Ervin FR: Tryptophan depletion causes a rapid

lowering of mood in normal males. Psychopharmacology 1985;87:173-177.

211- Zarate CA Jr, Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA,

Charney DS, Manji H: A randomized trial of an N-methyl-D-aspartate antagonist in

treatment-resistant major depression. Arch Gen Psychiatry 2006;63:856-864.

212- Zarate C Jr, Machado-Vieira R, HenterI, Ibrahim L, Diazgranados N, Salvadore G:

Glutamatergic modulators: the future of treating mood disorders? Harv Rev Psychiatry

2010;18:293-303.

213- Zigmond AS, Snaith RP: The hospital anxiety and depression scale. Acta Psychiatr

Scand 1983;67:361-370.

! <:?!

214- Zimmerman M, Posternak MA, Chelminski I: Derivation of a definition of remission

on the Montgomery-Asberg depression rating scale corresponding to the definition of

remission on the Hamilton rating scale for depression. J Psychiatr Res 2004;38:577-582.

215- Zink M, Vollmayr B, Gebicke-Haerter PJ, Henn FA: Reduced expression of GABA

transporter GAT3 in helpless rats, an animal model of depression. Neurochem Res

2009;34:1584-1593.

! <:D!

Appendix I

Informed consent

5%7&2.6%'.,!()!$%/8*8'%!&'/!V,1*+(-(K8*&-!$%/8*8'%I!Y'8L%2,8.1!()!N.&K(!

!

Rapid depression treatment in cancer patients/Information Sheet, Consent Form Ver 20/12/10

!

RAPID DEPRESSION TREATMENT IN PATIENTS WITH CANCER– INFORMATION SHEET FOR INTERESTED INDIVIDUALS

#

P+&'J! 1(0! )(2! ,+(H8'K! &'! 8'.%2%,.! 8'! .+8,! 72(Z%*.M! V-%&,%! 2%&/! .+8,! 8')(26&.8('! ,+%%.!

*&2%)0--1! G%)(2%! /%*8/8'K! H+%.+%2! (2! '(.! 8.! 8,! &772(728&.%! )(2! 1(0! .(! .&J%! 7&2.M! [(02!

7&2.8*87&.8('! 8,!%'.82%-1!L(-0'.&21M! Q)! 1(0!/%*8/%!'(.! .(! .&J%!7&2.I! 8.!H8--!'(.!&))%*.!&'1!

)0.02%! *&2%! (2! .2%&.6%'.M! Q)! 1(0! /(! &K2%%! .(! .&J%! 7&2.! 8'! .+%! ,.0/1I! 1(0! &2%! )2%%! .(!

H8.+/2&H!&.!&'1!.86%!H8.+(0.!+&L8'K!.(!K8L%!&!2%&,('!&'/!.+8,!H8--!8'!'(!H&1!&))%*.!&'1!

)0.02%!+%&-.+!*&2%M!P+%!)(--(H8'K!,+%%.!%\7-&8',!H+&.!.+%!,.0/1!H8--!8'L(-L%M!!

V-%&,%!2%&/!.+%!8')(26&.8('!8'!.+%!2%,.!()!.+8,!8')(26&.8('!,+%%.!*&2%)0--1M!

!

8690#12#06'#%1.#-4#06'#:)-;'<0=#

Rates of depression in patients with terminal cancer are high - up to 70% have clinically significant symptoms of depression, and up to 25% may meet criteria for major depression. Although antidepressant drugs can be effective in patients with terminal cancer, they are slow to work (there can be a delay of up to 6-8 weeks between starting dosing and mood improvement), and are therefore not that helpful to patients needing rapid relief. Having an antidepressant that lifts mood fast might be very helpful, especially to improve quality of life for patients with cancer and their families. Low doses of ketamine, a drug that blocks the brain chemical glutamate, can improve symptoms of depression rapidly (within 2 hours), for up to 7 days (see Figure). This effect can be sustained by use of repeated doses. Ketamine is commonly used to treat pain. The doses we plan to use in this study are similar to doses used for treating pain, and are safe and well tolerated. Ketamine has been used extensively in man for over 40 years. Ketamine was originally developed as an anaesthetic drug, and is also commonly used to treat pain. The doses used for anaesthesia are about 10 times greater that the doses used for treating pain. The doses we plan to use in this study are similar to those used to treat pain. We have local experience with ketamine in major depression and can report similar positive outcomes to those described above. !

]%!7-&'!.(!.%,.!.+%!&'.8/%72%,,&'.!%))%*.,!()!J%.&68'%!8'!D!,.&K%,U!!

C.&K%!<U!V&2.8*87&'.,!H8--!2%*%8L%!&!,8'K-%!8'.2&60,*0-&2!8'Z%*.8('!()!J%.&68'%!9<6K^JK>M!

]%!H8--!&,,%,,!*+&'K%,!8'!1(02!6((/!(L%2!.+%!'%\.!A!/&1,!

!

5%7&2.6%'.,!()!$%/8*8'%!&'/!V,1*+(-(K8*&-!$%/8*8'%I!Y'8L%2,8.1!()!N.&K(!

!

Rapid depression treatment in cancer patients/Information Sheet, Consent Form Ver 20/12/10

!

!

C.&K%! ?U! V&2.8*87&'.,! H+(! +&L%! ,+(H'! 8672(L%6%'.! 8'! .+%82! /%72%,,8('! *&'! 2%*%8L%!

)02.+%2! 8'.2&60,*0-&2! 8'Z%*.8(',! ()! J%.&68'%M! ]%! &'.8*87&.%! .+8,! H8--! G%! ('*%! H%%J-1!

9G&,%/!('!+(H!-('K!.+%!%))%*.,!()!J%.&68'%!-&,.>M!

C.&K%!DU!V&2.8*87&'.,!H8--!2%*%8L%!('%!()!.+2%%!/(,%!-%L%-,!()!J%.&68'%!9:M<I!:M"!(2!

<M:6K^JK>M!]%!H8--!&,,%,,!*+&'K%,!8'!1(02!6((/!(L%2!.+%!'%\.!A!/&1,M!

!

]%!H8--!&/L8,%!1(0!H+&.!,.&K%!H%!&2%!8'L8.8'K!1(0!.(!7&2.8*87&.%!8'M!

!

8690#7>?'#-4#:9)01<1?9502#9)'#@'#+--A15B#4-)=#

• $%'!&'/!H(6%'I!&K%/!G%.H%%'!?:;#"!1%&2,_!• [(0!*&'!+&L%!&'1!.17%!()!*&'*%2I!&'/!&!-8)%!%\7%*.&'*1!&.!-%&,.!D!6('.+,_!• [(0!60,.!G%!6%/8*&--1!,.&G-%_!• [(0!60,.! +&L%! ,8K'8)8*&'.! ,167.(6,! ()! /%72%,,8('! 9H%!H8--! 6%&,02%! .+8,! 0,8'K! &!

2&.8'K!,*&-%>M!

You can discuss your suitability to participate in more detail with your doctor/oncologist. What will Participants be Asked to Do? 5&1!<U!4.!.+%!)82,.!L8,8.I!H%!H8--!*(--%*.!/%.&8-,!&G(0.!1(0!&'/!1(02!*&'*%2M!]%!H8--!2&.%!

1(02!6((/M!`%.&68'%!H8--!G%!K8L%'!&,!&'!8'Z%*.8('!8'.(!1(02!G0..(*JM!4).%2!.+%!J%.&68'%!

8'Z%*.8('!H%!H8--!2&.%!1(02!6((/!,%L%2&-!.86%,!(0.!.(!@!+(02,I!&'/!2%*(2/!&'1!,8/%!

%))%*.,!1(0!68K+.!%\7%28%'*%M!!P+8,!7&2.!()!.+%!,.0/1!H8--!.&J%!&G(0.!@!a!!+(02,I!&'/!H8--!

(**02!8'!&'!(0.7&.8%'.!*-8'8*!8'!.+%!('*(-(K1!/%7&2.6%'.M!!

5&1!?I!DI!@I!=U!]%!H8--!&K&8'!2&.%!1(02!6((/!&'/!2%*(2/!&'1!,8/%!%))%*.,M!b&*+!()!.+%,%!

L8,8.,!H8--!-&,.!'(!6(2%!.+&.!<";?:!68'0.%,I!&'/!*(0-/!.&J%!7-&*%!&.!.+%!+(,78.&-!(2!&.!

1(02!+(0,%!9H+&.%L%2!8,!6(,.!*('L%'8%'.!)(2!1(0>M!P+%2%!6&1!G%!)%H%2!,*+%/0-%/!L8,8.,!

)(2!7&.8%'.,!%'2(--%/!8'!C.&K%!?I!H+%2%!2%7%&.%/!J%.&68'%!/(,%,!&2%!0,%/M!!

!

8690#%)'#76'#*1,'/C44'<02#%5,#D12A2=##

P+%!6(,.!*(66('!,8/%!%))%*.,!()!J%.&68'%!8'*-0/%!)%%-8'K!H((c1!8'!.+%!+%&/I!/2(H,1I!

&'/!/8cc1_!'06G'%,,!8'!.+%!)&*%!&'/!.('K0%_!+&L8'K!1(02!&26,!&'/!-%K,!)%%-8'K!+%&L1!(2!

'(.!*(''%*.%/!.(!1(0_!)%%-8'K!0',.%&/1!('!1(02!)%%.M!P+%,%!,8/%!%))%*.,!&2%!6(,.!

'(.8*%&G-%!&2(0'/!<:;?:!68'0.%,!&).%2!8'Z%*.8('I!&'/!&2%!K('%!&).%2!@:;#:!68'0.%,M!

!

B8,J,!8'*-0/%U!/8,*(6)(2.!)2(6!&'!8'.2&60,*0-&2!8'Z%*.8('_!.+%!,8/%!%))%*.,!)2(6!.+%!

J%.&68'%!8'Z%*.8('!9&,!/%,*28G%/!&G(L%>M!P+%!J%.&68'%!8'Z%*.8('!/(%,!'(.!8672(L%!6((/!

8'!,(6%!7&.8%'.,U!8)!.+8,!(**02,I!1(02!/(*.(2!*&'!/8,*0,,!&-.%2'&.8L%!.2%&.6%'.,!)(2!1(02!

/%72%,,8('!H8.+!1(0M!bL%'!8'!.+(,%!7&.8%'.,!H+%2%!.+%!J%.&68'%!+%-7,!-8).!1(02!

/%72%,,8('I!)02.+%2!8'Z%*.8('9,>!6&1!G%!'%%/%/I!&'/^(2!(.+%2!&'.8/%72%,,&'.!

.2%&.6%'.,M!

5%7&2.6%'.,!()!$%/8*8'%!&'/!V,1*+(-(K8*&-!$%/8*8'%I!Y'8L%2,8.1!()!N.&K(!

!

Rapid depression treatment in cancer patients/Information Sheet, Consent Form Ver 20/12/10

!

!

81++#E#F'#?91,#4-)#.>#?9)01<1?901-5#-5#0612#?)-;'<0=#

R(M!P+%!G%'%)8.,!H8--!*(6%!)2(6!.+%!.2%&.6%'.!72(L8/%/!G1!.+%!6%/8*8'%!0,%/!8'!.+%!

,.0/1M!

!

81++#10#F'#95>#<-202#4-)#.>#?9)01<1?901-5#-5#0612#?)-;'<0=##

R(M!Q.!H8--!G%!'(!*(,.,!)(2!1(02,%-)!(2!)&68-1!2%K&2/8'K!.+8,!72(Z%*.M!!

!

!95#:9)01<1?9502#!695B'#06'1)#G15,#95,#8106,)9@#4)-.#06'#:)-;'<0=#

[(0!6&1!H8.+/2&H!)2(6!.+8,!72(Z%*.!&.!&'1!.86%!&'/!H8.+(0.!&'1!/8,&/L&'.&K%!()!&'1!

J8'/M!!

!

8690#H909#-)#E54-).901-5#@1++#F'#!-++'<0',#95,#8690#I2'#@1++#F'#G9,'#-4#10=#

]%! &2%! *(--%*.8'K! 8')(26&.8('! &G(0.! *+&'K%,! 8'! 1(02!6((/! &).%2! J%.&68'%I! 8'*-0/8'K!

+(H!T08*J-1! .+8,!(**02,I!&'/!+(H! -('K! 8.! -&,.,! )(2M!]%!H8--! 2%*(2/!&'1!,8/%!%))%*.,!1(0!

2%7(2.I! &'/! (.+%2! ,&)%.1! 6%&,02%,! 9G-((/! 72%,,02%I! %.*>M! ]%! H8--! &'&-1,%! .+8,!

8')(26&.8('!,.&.8,.8*&--1!.(!/%.%268'%!.+%!)(--(H8'KU!!

• +(H!%))%*.8L%!J%.&68'%!8,!&,!&'!&'.8/%72%,,&'.I!!

• +(H!)&,.!8.!H(2J,I!!

• +(H!-('K!.+%!%))%*.,!-&,.!)(2I!&'/!

• H+&.!8,!.+%!G%,.!/(,%!.(!0,%M!

!

P+%!2%,0-.,!H8--!G%!70G-8,+%/!8'!&'!8'.%2'&.8('&-!,*8%'.8)8*!Z(02'&-M!P+%!/&.&!8'*-0/%/!8'!

.+8,!70G-8*&.8('!H8--!8'!'(!H&1!G%!-8'J%/!.(!&'1!,7%*8)8*!7&2.8*87&'.!&'/!1(02!8/%'.8.1!H8--!

'(.!G%!2%*(2/%/!H8.+! .+%!/&.&M! Q)!1(0!H&'.!&**%,,! .(!1(02!7%2,('&-!/&.&! -&.%2!1(0!H8--!

'%%/! .(! 2%*(2/! .+%! 8/%'.8)8*&.8('!'06G%2!0,%/! )(2! 1(02!7&2.8*0-&2! .%,.,M! [(0!&2%!6(,.!

H%-*(6%!.(!2%T0%,.!&!*(71!()!.+%!2%,0-.,!()!.+%!72(Z%*.!,+(0-/!1(0!H8,+M!

!

P+%!/&.&!*(--%*.%/!H8--!G%!,%*02%-1!,.(2%/!8'!,0*+!&!H&1!.+&.!('-1!.+(,%!8'L(-L%/!8'!.+%!

2%,%&2*+!72(K2&6!H8--!G%!&G-%!.(!K&8'!&**%,,!.(!8.M!!4.!.+%!%'/!()!.+%!72(Z%*.!&'1!7%2,('&-!

8')(26&.8('!H8--!G%!/%,.2(1%/!866%/8&.%-1!%\*%7.!.+&.I!&,!2%T082%/!G1!.+%!Y'8L%2,8.1d,!

2%,%&2*+!7(-8*1I!&'1!2&H!/&.&!('!H+8*+!.+%!2%,0-.,!()!.+%!72(Z%*.!/%7%'/!H8--!G%!2%.&8'%/!

8'!,%*02%!,.(2&K%!)(2!.%'!1%&2,I!&).%2!H+8*+!8.!H8--!G%!/%,.2(1%/M!

#

8690#14#>-J#69('#95>#KJ'201-52=#

Q)!1(0!+&L%!&'1!T0%,.8(',!&G(0.!(02!72(Z%*.I!%8.+%2!'(H!(2!8'!.+%!)0.02%I!7-%&,%!)%%-!)2%%!

.(!*('.&*.!0,!L8&!.+%!/%.&8-,!K8L%'!G%-(HM!

5%7&2.6%'.,!()!$%/8*8'%!&'/!V,1*+(-(K8*&-!$%/8*8'%I!Y'8L%2,8.1!()!N.&K(!

!

Rapid depression treatment in cancer patients/Information Sheet, Consent Form Ver 20/12/10

!

!-509<0#H'091+2U!!

V2()%,,(2!V&0-!e-0%I!5%7.!()!V,1*+(-(K8*&-!$%/8*8'%_!.%-!:D!@A:!D=#A_!%6&8-U!

7&0-MK-0%f(.&K(M&*M'c!

4,,(*8&.%!V2()%,,(2!5&L8/!V%2%cI!50'%/8'!V0G-8*!3(,78.&-_!.%-!:D@A@!:EEE_!%6&8-U!

/&L8/M7%2%cf,(0.+%2'/+GMK(L.M'c!

52!W-&0/8&!g&'8*(..8I!5%7.!()!V,1*+(-(K8*&-!$%/8*8'%_!.%-!:D!@A:!E@"<_!%6&8-U!

*-&0/8&K2(..f+(.6&8-M*(6!

%!!#2090'.'50L##

Q'!.+%!0'-8J%-1!%L%'.!()!&!7+1,8*&-!8'Z021!&,!&!2%,0-.!()!1(02!7&2.8*87&.8('!8'!.+8,!,.0/1I!

1(0!H8--!G%!*(L%2%/!G1!.+%!&**8/%'.!*(67%',&.8('!-%K8,-&.8('!H8.+!8.,!-868.&.8(',M!Q)!1(0!

+&L%!&'1!T0%,.8(',!&G(0.!4WW!7-%&,%!)%%-!)2%%!.(!&,J!.+%!2%,%&2*+%2!)(2!6(2%!

8')(26&.8('!G%)(2%!1(0!&K2%%!.(!.&J%!7&2.!8'!.+8,!.28&-M!

!

M'9+06#95,#,129F1+10>#2090'.'50L#

Q)!1(0!+&L%!&'1!T0%28%,!(2!*('*%2',!&G(0.!1(02!28K+.,!&,!&!7&2.8*87&'.!8'!.+8,!,.0/1!1(0!

6&1!H8,+!.(!*('.&*.!&!3%&-.+!&'/!58,&G8-8.1!C%2L8*%,!W(',06%2!4/L(*&.%I!.%-%7+('%U!

9:D>!@AE!:?#"!(2!)2%%7+('%!:=::!DA!AA!##!(2!)2%%)&\!:=::!?A=A!A#A=!9:=::!?!

CYVVNBP>!(2!%6&8-!&/L(*&*1f+/*M(2KM'c!Q)!.+%2%!8,!&!,7%*8)8*!$&(28!8,,0%^*('*%2'!

7-%&,%!*('.&*.!S8'/&!e2%''%--!&.!:=::!DAA!A##M!

!

P+8,!72(Z%*.!+&,!G%%'!2%L8%H%/!&'/!&772(L%/!G1!.+%!S(H%2!C(0.+!B%K8('&-!b.+8*,!

W(668..%%!9SBC^<:^<<^:":>M!

5%7&2.6%'.,!()!$%/8*8'%!&'/!V,1*+(-(K8*&-!$%/8*8'%I!Y'8L%2,8.1!()!N.&K(!

!

Rapid depression treatment in cancer patients/Information Sheet, Consent Form Ver 20/12/10

!

!

RAPID DEPRESSION TREATMENT IN PATIENTS WITH CANCER– #

!3&*C&7#N3DG#N3D#:%D7E!E:%&7*#

#

I have read the “RAPID DEPRESSION TREATMENT IN PATIENTS WITH CANCER” Information Sheet concerning this project. All my questions have been answered to my satisfaction. I understand that I am free to request further information at any stage. !

Q!J'(H!.+&.U!

1. I am free to withdraw from testing at any time without any disadvantage; ?M Q!0'/%2,.&'/!.+%!/&.&!H8--!G%!/%,.2(1%/!&.!.+%!*('*-0,8('!()!.+%!72(Z%*.!G0.!&'1!

2&H!/&.&!('!H+8*+!.+%!2%,0-.,!()!.+%!72(Z%*.!/%7%'/!H8--!G%!2%.&8'%/!8'!,%*02%!

,.(2&K%!)(2!.%'!1%&2,I!&).%2!H+8*+!8.!H8--!G%!/%,.2(1%/_!

3. I understand the results of the project may be published but my anonymity will be preserved.

I agree to take part in this project. ………………………………………………… (Full name) ............................................................................ ............................... (Signature) (Date) ………………………………………………. (Full name) ……………………………………………….. ……………………. (Doctor’s / Researcher Signature) (Date) P+8,!72(Z%*.!+&,!G%%'!2%L8%H%/!&'/!&772(L%/!G1!.+%!S(H%2!C(0.+!B%K8('&-!b.+8*,!W(668..%%!

9SBC^<:^<<^:":

!

!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!

!!

!!!!!<:E!

!

Appendix II

Case report – in press (page proofs)

!

!

Brief Reports

Mood and Pain Responses to Repeat Dose IntramuscularKetamine in a Depressed Patient with Advanced Cancer

Claudia G. Zanicotti, M.D.,1 David Perez, M.D., FRACP,2 and Paul Glue, M.D., FRCPsych1

Abstract

Depression is highly prevalent in patients with advanced cancer, commonly affecting quality of life. Consideringthe response delay with conventional antidepressants and the short life expectancy for these patients, treatmentsfor Major Depressive Disorder (MDD) with faster onset of action are desirable. In this case report, a femalepatient with metastatic ovarian cancer reported rapid and sustained response to intramuscular (IM) injections ofketamine (1mg/kg). Over a course of six treatments, her mood response was identical on each occasion andprovided remission of her depressive symptoms. Pain was also improved, although for a shorter duration. Thesefindings support the use of IM ketamine as a possible antidepressant option for this population.

Introduction

The prevalence ofMajor Depressive Disorder (MDD) inadult patients with cancer is high. In those receiving

palliative care the rate is considered approximately 15%,1,2

and the rate of severe depression in advanced cancer patientsis around 11%.3

Current antidepressants target the monoaminergic systemand typically have a slow onset of action. More recently, therehas been an awareness of the role of the glutamatergic systemin the pathophysiology of mood disorders.4 Ketamine – anoncompetitive NMDA (N-Methyl-D-Aspartate) antagonist –has recently been identified as a possible fast-acting treatmentfor MDD.5,6 Moreover, the use of ketamine for pain in ad-vanced cancer patients might also pose an extra benefit as anadjuvant therapy.7

The following case report describes a patient enrolled inan ongoing clinical trial, which was approved by theLower South Regional Ethics Committee, Southern HealthDistrict Board and Dunedin School of Medicine, Universityof Otago, New Zealand. This trial is registered (ACTRN12610001011077).

The patient participated in Stages 1 and 2 of this trial. Stage1 consists of a single open label injection of IM ketamine 1mg/kg. Patients whose mood improved, with Montgomery-Asberg Depression Rating Scale (MADRS)8 score reduced by50% or more, could proceed to Stage 2. This stage consists ofopen label characterization of safety and efficacy of repeatdose IM ketamine, administered at approximately weeklyintervals.

Inclusion criteria are diagnosis of MDD (DSM IV-TR);9

MADRS > 20; presence of incurable cancer; written informedconsent; life expectancy > 3 months; age between 20 and 65years old; ECOG bAU1status < 4; and adequate hepatic, renal, andhematological function. Exclusion criteria are current use ofketamine; presence of psychosis or delirium; presence of ce-rebral metastases; Comparative Pain Scale10 score of > 5; andany antidepressant treatment started within the previous4 weeks.

Case Report

A. is a 36-year-old female patient with metastatic ovariancarcinoma. She has had MDD and Dysthymic Disorder di-agnosed since she was 18 years old. Even though she has triedvarious antidepressants, the patient reports that her moodwas never completely improved. For the past 10 years she hada worsening of her depressive symptoms, which persisted atthe time of her first assessment, and included low mood, lackof energy, irritability, easy crying, sleep disturbance, lack ofconcentration, excessive feelings of guilt, and passive deathwish. Her domestic circumstances were stressful, due toconcern about the future of her 11-year-old daughter and thelack of support from her ex-boyfriend (the child’s father) andher family.

Along with depression, A. has had visceral pain for *8months in the right iliac fossa, in the same location as a met-astatic tumor, which is about 7cm in diameter on palpation.For pain intensity assessment, The Comparative Pain Scale,10

with a range from 0 (no pain) to 10 (short unconsciousness),

1Department of Psychological Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand.2Department of Oncology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand.Accepted: November 18, 2011.

JOURNAL OF PALLIATIVE MEDICINEVolume 15, Number 4, 2012ª Mary Ann Liebert, Inc.DOI: 10.1089/jpm.2011.0314

1

JPM-2011-0314-ver9-Zanicotti_1P.3d 03/26/12 2:04pm Page 1

JPM-2011-0314-ver9-Zanicotti_1P

!

!

was used. She also has a previous history of drug abuse(methamphetamine), being abstinent for more than six years,and a family history of alcoholism (both grandfathers andone uncle).

When first assessed for ketamine treatment, A. was takingvenlafaxine 150mg/day, quetiapine 50mg/day, and metha-done 95mg/day. All her medications were continued duringthe ketamine treatment with doses unchanged. She was alsoreceiving weekly chemotherapy (combination of carboplatinand gemcitabine). After informed consent was given, she re-ceived ketamine 1mg/kg IM open-label. Dosing was per-formed on an outpatient basis in a hospital clinic.

MADRS was used as the main scale to assess her depres-sion scores. This is a validated scale that includes most DSM-IV criteria for MDD.8 It consists of 10 items, each one scoredfrom 0 to 6 (total range from 0 to 60). Scores > 20 are standardfor entry into antidepressant clinical trials.6,11 Mood and painassessments were obtained at baseline (20 minutes before in-jection), 1, 2, 4, 24, 48, 72, and 168 hours post-injection. TheHospital Anxiety and Depression Scale (HADS)12,13 is oftenused in patients with cancer or other somatic diseases, and itsoptimal cut-off scores are > 7 on both HADS-A and HADS-Dfor anxiety and depression respectively. There are no physicalsymptoms in its assessment, lessening the effects of physicalillness over depression and anxiety symptoms.13 The De-moralization Scale (DS)14 consists of 24 questions (total scorerange = 0 to 96), and a score > 30 is considered high. HADSand DS were assessed at baseline and 24 hours.

The patient’s initial MADRS score was 24, which droppedto 7 one hour after the first dose of ketamine. For the next twodays herMADRS remained low (5 points), with improvementin all MADRS scale items. Her depressive symptoms returnedafter 6 to 7 days post-injection. Also, her pain score was re-duced within a few minutes after the first ketamine dose,however returned to baselinewithin 24 hours (F1c Figure 1A). Shereported moderate but tolerable dissociative side effects, suchas depersonalization and immobility, that started 5 minutespost-injection and which resolved within 45 to 60 minutes.Vital signs were unchanged.

The patient has subsequently received repeat ketaminedoses at intervals of 7 to 8 days, based on return of her de-pressive symptoms. Her mood response to repeat injectionshas been identical each time, in terms of speed andmagnitudeof response (Figure 1B). Onset of pain relief continued to berapid, however the durability of analgesic response has beenmuch more variable (Figure 1C). Reported side effects weresimilar in timing of onset and intensity to those noted after thefirst dose. Scores on HADS and DS showed consistent im-provement in her mood symptoms ( bT1Table 1).

The improvement in the patient’s mood was noticed by herfamily, even on the phone while talking to her sister. Also, herdaughter said she was calmer and more patient. In visits toA.’s house to collect mood assessments these reported find-ings were verified.

Discussion

This is the second case report of ketamine being used as afast acting antidepressant in patients with cancer.15 Thepresent report extends information on ketamine use by as-sessing simultaneous effects on mood and pain ratings, andby demonstrating the reproducibility of the antidepressantresponse with repeated dosing.

The patient had remission of her depressive symptomswithin one hour after receiving the first injection of IM keta-mine (MADRS < 10).16 This is consistent with previous re-search that noted a rapidmood response for more than 70% ofpatients within 24 hours.17,18 The patient’s improvement wassustained for 6 to 7 days, with a gradual return to baseline inher depression scores, which is also consistent with the liter-ature.18,19 Some authors have noticed that there are patientswith a mood response sustained for even longer periods oftime.18,20 Most previous studies have used an IV infusion over40 minutes rather than IM injections, as used in this case.Pharmacokinetic simulations suggest comparable exposuresafter IM injection and IV infusion.21 The dose used in this case(1mg/kg) is higher than the dose used in most publishedcases (0.5mg/kg),17,18,19 and the optimal dose for treating

FIG. 1A. Change in depression ratings (!) and pain score (r) after first dose of IM ketamine. 1B: Reproducible changes indepression ratings after repeated ketamine administration - doses 1(!), 2(;), 3(-), 4(r), 5(:), 6( ). 1C: Variable changes inpain ratings after repeated administration (legend as for 1(B)). 1D: Correlation of change from baseline in MADRS and painratings (Pearson’s r) by time post dose.

JPM-2011-0314-ver9-Zanicotti_1P.3d 03/26/12 2:04pm Page 2

2 ZANICOTTI ET AL.

!

!

depression is still unclear. Changes in the DS and HADSratings were consistent with the MADRS data.

The analgesic effects were also fast in onset, however thedurability was variable after day one. Changes in pain scoresand depression ratings were most closely correlated 1 hourpost-dosing (Pearson’s r = 0.61) with progressively weakercorrelations over the next 168 hours (Figure 1D). This suggeststhat the sustained antidepressant effects of ketamine are me-diated differently from the mechanisms associated with im-mediate changes in pain and mood. One possible mechanismmay be via stimulation of mTOR signaling and via synapticprotein synthesis.22

Our patient had no tachyphylaxis, as demonstrated by theconsistent improvement in her mood scores at every injection.Messer and colleagues20 reported similar findings, withmaintenance of remission in one patient for more than 15months, with ketamine infusions given every three weeks. Itshould be noted that Liebrenz and colleagues23 reported adifferent response to a second ketamine infusion in one pa-tient, with worsening of depressive symptoms two days aftera second injection. This suggests that there may be individualdifferences in mood responses to repeated doses of ketamine.

According to Phelps and colleagues,24 the patient’s positivemood response might be related to her family history of al-coholism, affecting two of her second-degree relatives. In theabove mentioned study the authors compared ketamine re-sponse in two groups of depressed patients, one with familyhistories of alcoholism and the second without. Their resultssuggest that a family history of alcohol dependence may be amarker for mood response to an NMDA antagonist but, asthis was the first study with depressed patients, it requiresfurther studies for confirmation.

Administration by IM injection may have benefits whencompared to IV infusion, such as faster administration, withless discomfort to the patient; and reduced costs, without theneed of a pump for the infusion. These aspects, together withher positive outcome, support the use of IM injections in fu-ture research.

The use of a 1mg/kg IM dose was well tolerated andachieved remission of MDD. However, the optimal dose hasnot yet been established.21 Further data are required to de-termine optimal use of ketamine as a fast-acting antidepres-

sant. Considering that antidepressants available today have adelay in their response of usually 4 to 6 weeks, these findingsmay justify the use of IM ketamine as an antidepressant agentfor a population with a short life expectancy, as described inthis case report.

Acknowledgments

This research was supported by Otago Medical ResearchFoundation Inc. and the Otago Community Trust.

Authors Disclosure Statement

All authors state no competing financial interests exist.

References

1. Hotopf M, Chidgey J, Addington-Hall J, Ly KL: Depressionin advanced disease: A systematic review. Part 1. Prevalenceand case finding. J Palliat Med 2002;16:81–97.

2. Mitchell AJ, Chan M, Bhatti H, Halton M, Grassi L, JohansenC, Meader N: Prevalence of depression, anxiety, and ad-justment disorder in oncological, haematological, andpalliative-care settings: A meta-analysis of 94 interview-based studies. Lancet Oncol 2011;12:160–174.

3. Salvo N, Zeng L, Zhang L, Leung M, Khan L, Presutti R,Nguyen J, Holden L, Culleton S, Chow E: Frequency of re-porting and predictive factors for anxiety and depression inpatients with advanced cancer. Clin Oncol (R Coll Radiol)2011;6:in press.

4. Hashimoto K: Emerging role of glutamate in the patho-physiology of major depressive disorder. Brain Res Rev2009;61:105–123.

5. Zarate C Jr, Machado-Vieira R, Henter I, Ibrahim L, Dia-zgranados N, Salvadore G: Glutamatergic modulators: Thefuture of treating mood disorders? Harv Rev Psychiatry2010;18:293–303.

6. Machado-Vieira R, Yuan P, Brutsche N, Diazgranados N,Luckenbaugh D, Manji HK, Zarate CA Jr: Brain-derivedneurotrophic factor and initial antidepressant response to anN-methyl-D-aspartate antagonist. J Clin Psychiatry 2009;70:1662–1666.

7. Mercadante S, Porzio G, Gebbia V: Spinal analgesia for ad-vanced cancer patients: An update. Crit Rev Oncol Hematol2011;16:in press.

8. Montgomery SA, Asberg M: A new depression scale de-signed to be sensitive to change. Br J Psychiatry 1979;134:382–389.

9. American Psychiatric Association: Diagnostic and StatisticalManual of Mental Disorders (4th ed). Washington, DC,2000..

10. Harich, J: The comparative pain scale. www.tipna.org/info/documents/ComparativePainScale.htm. (Last acessed 10/03/2011.)

11. Diazgranados N, Ibrahim L, Brutsche NE, Newberg A,Kronstein P, Khalife S, Kammerer WA, Quezado Z, Luck-enbaugh DA, Salvadore G, Machado-Vieira R, Manji HK,Zarate CA Jr: A randomized add-on trial of an N-methyl-D-aspartate antagonist in treatment-resistant bipolar depres-sion. Arch Gen Psychiatry 2010;67:793–802.

12. Bjelland I, Dahl AA, Haug TT, Neckelmann D: The validityof the Hospital Anxiety and Depression Scale. An updatedliterature review. J Psychosom Res 2002;52:69–77.

13. Zigmond AS, Snaith RP: The hospital anxiety and depres-sion scale. Acta Psychiatr Scand 1983;67:361–370.

Table 1: Assessment of HADSand Demoralization Scale

Dose Assessments Baseline24 hours

post-injection

Dose 1 HADS 23 (D:14 A:9) 5 (D:3 A:2)DS 71 29

Dose 2 HADS 27 (D:17 A:10) 4 (D:2 A:2)DS 78 28

Dose 3 HADS 25 (D:18 A:7) 3 (D:2 A:1)DS 77 28

Dose 4 HADS 23 (D:15 A:8) 6 (D:6 A:0)DS 82 32

Dose 5 HADS 22 (D:16 A:6) 8 (D:6 A:2)DS 77 35

Dose 6 HADS 26 (D:15 A:11) 6 (D:5 A:1)DS 78 26

DS, Demoralization Scale; D, HADS depression scores; A, HADSanxiety scores.

JPM-2011-0314-ver9-Zanicotti_1P.3d 03/26/12 2:04pm Page 3

KETAMINE FOR DEPRESSION IN A CANCER PATIENT 3

!

!

14. Kissane DW, Wein S, Love A, Lee XQ, Kee PL, Clarke DM:The Demoralization Scale: A report of its development andpreliminary validation. J Palliat Care 2004;20:269–276.

15. Stefanczyk-Sapieha L, Oneschuk D, Demas M: Intravenousketamine ‘‘burst’’ for refractory depression in a patient withadvanced cancer. J Palliat Med 2008;11:1268–1271.

16. Hawley CJ, Gale TM, Sivakumaran T: Defining remission bycut off score on the MADRS: Selecting the optimal value. JAffect Disord 2002;72:177–184.

17. Zarate CA Jr, Singh JB, Carlson PJ, Brutsche NE, Ameli R,Luckenbaugh DA, Charney DS, Manji HK: A randomized trialof an N-methyl-D-aspartate antagonist in treatment-resistantmajor depression. Arch Gen Psychiatry 2006;63:856–864.

18. Aan het Rot M, Collins KA, Murrough JW, Perez AM, ReichDL, Charney DS, Mathew SJ: Safety and efficacy of repeated-dose intravenous ketamine for treatment-resistant depres-sion. Biol Psychiatry 2010;67:139–145.

19. Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR,Charney DS, Krystal JH: Antidepressant effects of ketaminein depressed patients. Biol Psychiatry 2000;47:351–354.

20. Messer MM, Haller IV: Maintenance ketamine treatment pro-duces long-term recovery from depression. Primary Psychiatry2010;17:48–50.

21. Glue P, Gulati A, Le Nedelec M, Duffull F: Dose- andexposure-response to ketamine in depression. Biol Psy-chiatry 2011;7:in press.

22. Duman RS, Li N, Liu RJ, Duric V, Aghajanian G: Signalingpathways underlying the rapid antidepressan actions ofketamine. Neuropharmacology 2012;62:35–41.

23. Liebrenz M, Stohler R, Borgeat A: Repeated intravenousketamine therapy in a patient with treatment-resistant majordepression. World J Biol Psychiatry 2009;10:640–643.

24. Phelps LE, Brutsche N, Moral JR, Luckenbaugh DA, ManjiHK, Zarate CA Jr: Family history of alcohol dependence andinitial antidepressant response to an NMDA antagonist. BiolPsychiatry 2009;65:181–184.

Address correspondence to:Claudia Grott Zanicotti

Department of Psychological MedicineUniversity of Otago

PO Box 913Dunedin, New Zealand

E-mail: [email protected]

JPM-2011-0314-ver9-Zanicotti_1P.3d 03/26/12 2:04pm Page 4

4 ZANICOTTI ET AL.