Drug therapy as an intervention for Autism or Asperger's syndrome and other Autism Spectrum Disorders
 
 

ANTIDEPRESSANTS

An antidepressant is a psychiatric medication or other substance (nutrient or herb) used for alleviating depression or dysthymia ('milder' depression). Drug groups known as MAOIs, tricyclics and SSRIs are particularly associated with the term. These medications are now amongst the drugs most commonly prescribed by psychiatrists and general practitioners, and their effectiveness and adverse effects are the subject of many studies and competing claims. Nutrients for which there are claims of antidepressant activity include phenylalanine, tyrosine, tryptophan, 5-Hydroxytryptophan, and choline.

 

Most antidepressants have a delayed onset of action and are usually taken over the course of weeks, months or years. They are generally considered distinct from stimulants, and drugs used for an immediate euphoric effect only are not generally considered antidepressants. Despite the name, antidepressants are often used in the treatment of other conditions, including anxiety disorders, bipolar disorder, obsessive compulsive disorder, eating disorders and chronic pain. Some have also become known as lifestyle drugs or "mood brighteners". Other medications not known as antidepressants, including antipsychotics in low doses[1] and benzodiazepines,[2] are also widely used to manage depression.

 

The term antidepressant is sometimes applied to any therapy (e.g. psychotherapy, electro-convulsive therapy, acupuncture) or process (e.g. sleep disruption, increased light levels, regular exercise) found to improve clinically depressed mood. An inert placebo tends to have a significant antidepressant effect, so establishing something as an antidepressant in a clinical trial involves demonstrating a significant additional effect.

 

History

Saint John's Wort

Opium[3] and St John's Wort[4] (as a "nerve tonic") had long been used to alleviate depression, but the contemporary history of antidepressant medications begins with isoniazid.

 

Isoniazid and iproniazid

In 1951, two physicians from the Sea View Hospital on Staten Island, Irving Selikoff and Edward Robitzek, began clinical trials to evaluate two new anti-tuberculosis agents from Hoffman-LaRoche, isoniazid and iproniazid. Only the patients with poor prognosis were initially treated; nevertheless, their condition improved dramatically. In addition, Selikoff and Robitzek noted "a subtle general stimulation... The patients exhibited renewed vigor and indeed this occasionally served to introduce disciplinary problems."[5] The promise of the cure for tuberculosis brought by the results of the Sea View Hospital trials was also excitedly discussed in the mainstream press. In 1952, learning of the stimulant-like side effects of the isoniazid, the Cincinnati psychiatrist Max Lurie decided to try it on his patients. In the following year, he and Harry Salzer reported that isoniazid improved the depression in two thirds of their patients and also coined the term antidepressant to describe its action.[6] A similar story happened in Paris, where Jean Delay, the head of psychiatry at Sainte-Anne Hospital, found out from his pulmonology colleagues from Cochin Hospital about the side effects of isoniazid. In 1952, that is even earlier than Lurie and Salzer, Delay with the resident Jean-Francois Buisson also reported the positive action of isoniazid on depressed patients.[7] For the reasons unrelated to the efficacy, isoniazid as antidepressant was soon overshadowed by more toxic iproniazid,[6] although it remains one of the mainstays of the tuberculosis treatment. The mode of antidepressant action of isoniazid is still unclear. It is speculated that its effect is due to the inhibition of diamine oxidase coupled with a weak inhibition of monoamine oxidase A.[8]

 

Another anti-tuberculosis drug tried at the same time by Selikoff and Robitzek, iproniazid, was observed to have a greater "psychostimulant" effect, albeit at the cost of a greater toxicity.[9] Subsequently to the publications on isoniazid, papers by Jackson Smith, Gordon Kamman, George Crane and Frank Ayd describing the psychiatric applications of iproniazid also appeared, and Ernst Zeller found iproniazid to be a potent monoamine oxidase inhibitor.[10] Nevertheless, iproniazid had remained relatively obscure until Nathan Kline, the influential and flamboyant head of research at Rockland State Hospital, began its popularization both in medical and popular press as a "psychic energizer".[11][10] While isoniazid was not patentable,[6] Roche put a significant marketing effort behind iproniazid, including promoting its off-label use for depression.[10] Its sales grew massively in the following years, until it was recalled from the market in 1961 due to the cases of lethal hepatotoxicity.[10]

 

Imipramine

The discovery that a tricyclic ("three ringed") compound had a significant antidepressant effect was also first made in the early 1950s, by Roland Kuhn in a Swiss psychiatric hospital. By that time antihistamine derivatives were coming in to use to treat surgical shock and then as psychiatric neuroleptics. Although, in 1955, reserpine was indicated to be more effective than placebo in alleviating anxious depression, neuroleptics (literally "to seize the neuron") were developing for use as sedatives and antipsychotics.

 

In attempting to improve the effectiveness of one of them, chlorpromazine, in conjunction with the Geigy pharmaceutical company, Kuhn discovered that compound "G 22355" (manufactured and patented in the US in 1951 by Häfliger and Schinder) had a beneficial effect in patients with depression with mental and motor retardation.[12] He first reported his findings on what he called a "thymoleptic" (literally "taking hold of the emotions", by contrast with neuroleptics, "taking hold of the nerves") in 1955/56 and they gradually became established, resulting in the marketing of the first tricyclic antidepressant, imipramine, soon followed by variants.

 

Later history

These new drug therapies became prescription-only medications in the 1950s. It was estimated that no more than 50 to 100 people per million suffered from the kind of depression that these new drugs would treat and pharmaceutical companies were not enthusiastic. Sales through the 1960s remained poor compared to the major tranquilizers (neuroleptics/antipsychotics) and minor tranquilizers (such as benzodiazepines), which were being marketed for different uses.

 

The term antidepressant is reported to have been coined by Lurie and to not have been widely adopted until at least the 1960s.[13] Imipramine remained in common use and numerous successors were introduced. The field of MAO inhibitors remained quiet for many years until "reversible" forms affecting only the MAO-A subtype were introduced, avoiding some of the adverse effects.[14][15][16]

 

Most pharmacologists by the 1960s thought the main therapeutic action of tricyclics was to inhibit norepinephrine reuptake, but it was gradually observed that this action was associated with energizing and motor stimulating effects whilst some antidepressant compounds appeared to have differing effects through action on serotonin systems (notably proposed by Carlsson and Lindqvist (1969) and Lapin and Oxenkrug (1969)).

 

Researchers began a process of rational drug design to isolate antihistamine-derived compounds that would 'selectively' (specifically) target these systems. The first such compound to be patented, in 1971, was zimelidine, whilst the first released clinically was indalpine. Fluoxetine (Prozac), FDA approved for commercial use in 1988, became the first blockbuster SSRI. Fluoxetine was developed at Eli Lilly in the early 1970s by Bryan Molloy, David Wong and others.[17][18]

 

While it had fallen out of favor in most countries through the 19th and 20th centuries, the herb St John's Wort had become increasingly popular in Germany where Hypericum extracts eventually became licensed, packaged and prescribed by doctors. Small-scale efficacy trials were carried out from the 1970s and 1980s, and attention grew in the 1990s following a meta-analysis of these.[19] It remained an over-the-counter drug (OTC) or supplement in most countries and research continued to investigate its neurotransmitter effects and active components, particularly hyperforin[20][21]

 

SSRIs became known as "novel antidepressants" along with other newer drugs such as SNRIs and NRIs with various different selective effects, such as venlafaxine, duloxetine, nefazodone and mirtazapine[22]

 

Types of Antidepressants

Selective serotonin reuptake inhibitors (SSRIs)

Selective serotonin reuptake inhibitors (SSRIs) are a family of antidepressants considered to be the current standard of drug treatment. It is thought that one cause of depression is an inadequate amount of serotonin, a chemical used in the brain to transmit signals between neurons. SSRIs are said to work by preventing the reuptake of serotonin by the presynaptic nerve, thus maintaining higher levels of 5-HT in the synapse. Recently, however, work by two researchers has called into question the link between serotonin deficiency and symptoms of depression, noting that the efficacy of SSRIs as treatment does not in itself prove the link.[23] Recent research indicates that these drugs may interact with transcription factors known as "clock genes",[24] which may be important for the addictive properties of drugs of abuse, and possibly in obesity.[25][26]

 

Recent randomized controlled trials in Archives of General Psychiatry showed that up to one-third of effects of SSRI Treatment can be seen in first week. Early effects also shown to have secondary effect of reducing absolute reduction in HDRS score by 50%. Even more recent studies, published by the Archives of General Psychiatry note that 25% of so-called clinical depression does not meet a disease criteria and should be considered to be ordinary sadness and adjustment to the difficulties in life.

 

This family of drugs includes fluoxetine (Prozac), paroxetine (Paxil), escitalopram (Lexapro, Esipram), citalopram (Celexa), and sertraline (Zoloft). These antidepressants typically have fewer adverse side effects than the tricyclics or the MAOIs, although such effects as drowsiness, dry mouth, nervousness, anxiety, insomnia, decreased appetite, and decreased ability to function sexually may occur. Some side effects may decrease as a person adjusts to the drug, but other side effects may be persistent. Though safer than first generation antidepressants, SSRI's may not work as often, suggesting the role of norepinephrine.

 

Serotonin-norepinephrine reuptake inhibitors (SNRIs)

Serotonin-norepinephrine reuptake inhibitors (SNRIs) such as venlafaxine (Effexor) and duloxetine (Cymbalta) are a newer form of antidepressant that works on both norepinephrine and 5-HT. They typically have similar side effects to the SSRIs, although there may be a withdrawal syndrome on discontinuation that may necessitate dosage tapering.

 

Noradrenergic and specific serotonergic antidepressants (NASSAs)

Noradrenergic and specific serotonergic antidepressants (NASSAs) form a newer class of antidepressants which purportedly work to increase norepinephrine (noradrenaline) and serotonin neurotransmission by blocking presynaptic alpha-2 adrenergic receptors while at the same time minimizing serotonin related side-effects by blocking certain serotonin receptors. The only example of this class in clinical use is mirtazapine (Avanza, Zispin, Remeron).

 

Norepinephrine (noradrenaline) reuptake inhibitors (NRIs)

Norepinephrine (noradrenaline) reuptake inhibitors (NRIs) such as reboxetine (Edronax) act via norepinephrine (also known as noradrenaline). NRIs are thought to have a positive effect on concentration and motivation in particular, though they have been known to increase aggression.

 

Norepinephrine-dopamine reuptake inhibitors

Norepinephrine-dopamine reuptake inhibitors such as bupropion (Wellbutrin, Zyban) inhibit the neuronal reuptake of dopamine and norepinephrine (noradrenaline).[27]

 

Tricyclic antidepressants (TCAs)

Tricyclic antidepressants are the oldest and include such medications as amitriptyline and desipramine. Tricyclics block the reuptake of certain neurotransmitters such as norepinephrine (noradrenaline) and serotonin. They are used less commonly now due to the development of more selective and safer drugs. Several side effects include increased heart rate, drowsiness, dry mouth, constipation, urinary retention, blurred vision, dizziness, confusion, and sexual dysfunction. Toxicity occurs at approximately ten times normal dosages. However, tricyclic antidepressants are still used because of their high potency, especially in severe cases of clinical depression.

 

Monoamine oxidase inhibitor (MAOIs)

Monoamine oxidase inhibitors (MAOIs) such as phenelzine (Nardil) may be used if other antidepressant medications are ineffective. Because there are potentially fatal interactions between this class of medication and certain foods (particularly those containing Tyramine), as well as certain drugs, classic MAOIs are rarely prescribed anymore. MAOIs work by blocking the enzyme monoamine oxidase which breaks down the neurotransmitters dopamine, serotonin, and norepinephrine (noradrenaline). MAOIs can be as effective as tricyclic antidepressants, although they can have a higher incidence of dangerous side effects (as a result of inhibition of cytochrome P450 in the liver). A new generation of MAOIs has been introduced; moclobemide (Manerix), known as a reversible inhibitor of monoamine oxidase A (RIMA), acts in a more short-lived and selective manner and does not require a special diet. Additionally, (selegiline) marketed as Emsam in a transdermal form is not a classic MAOI in that at moderate dosages it tends to effect MAO-B which does not require any dietary restrictions.

 

Augmenter drugs

Some antidepressants have been found to work more effectively in some patients when used in combination with another drug. Such "augmenter" drugs include tryptophan (Tryptan) and buspirone (Buspar).

 

Tranquillizers and sedatives, typically the benzodiazepines, may be prescribed to ease anxiety and promote sleep. Because of their high potential for fostering dependence, these medications are intended only for short-term or occasional use. Medications often are used not for their primary function but to exploit what are normally side effects. Quetiapine fumarate (Seroquel) is designed primarily to treat schizophrenia and bipolar disorder, but a frequently reported side-effect is somnolence. Therefore, this drug can be used in place of an antianxiety agent such as clonazepam (Klonopin, Rivotril).

 

Antipsychotics such as risperidone (Risperdal), olanzapine (Zyprexa), and quetiapine (Seroquel) are prescribed as mood stabilizers and are also effective in treating anxiety. Their use as mood stabilizers is a recent phenomenon and is controversial with some patients. Antipsychotics (typical or atypical) may also be prescribed in an attempt to augment an antidepressant, to make antidepressant blood concentration higher, or to relieve psychotic or paranoid symptoms often accompanying clinical depression. However, they may have serious side effects, particularly at high dosages, which may include blurred vision, muscle spasms, restlessness, tardive dyskinesia, and weight gain.

 

Antidepressants by their nature behave similarly to psychostimulants. Antianxiety medications by their nature are depressants. Close medical supervision is critical to proper treatment if a patient presents with both illnesses because the medications tend to work against each other.

 

Psycho-stimulants are sometimes added to an antidepressant regimen if the patient suffers from anhedonia, hypersomnia and/or excessive eating as well as low motivation. These symptoms which are common in atypical depression can be quickly resolved with the addition of low to moderate dosages of amphetamine or methylphenidate (brand names Adderall and Ritalin, respectively) as these chemicals enhance motivation and social behavior, as well as suppress appetite and sleep. These chemicals are also known to restore sex drive. Extreme caution must be used however with certain populations. Stimulants are known to trigger manic episodes in people suffering from bipolar disorder. They are also easily abused as they are effective substitutes for Methamphetamine when used recreationally. Close supervision of those with substance abuse disorders is urged. Emotionally labile patients should avoid stimulants, as they exacerbate mood shifting.

 

Lithium remains the standard treatment for bipolar disorder and is often used in conjunction with other medications, depending on whether mania or depression is being treated. Lithium's potential side effects include thirst, tremors, light-headedness, and nausea or diarrhea. Some of the anticonvulsants, such as carbamazepine (Tegretol), sodium valproate (Epilim), and lamotrigine (Lamictal), are also used as mood stabilizers, particularly in bipolar disorder.

 

Prescription trends

In the United Kingdom the use of antidepressants increased by 234% in the 10 years up to 2002.[28] In the United States a 2005 independent report stated that 11% of women and 5% of men in the non-institutionalized population (2002) now take antidepressants[29] A 1998 survey found that 67% of patients diagnosed with depression were prescribed an antidepressant.[30] A 2007 study purports that 25% of Americans were overdiagnosed with depression, regardless of any medical intervention.[31] The findings were based on a national survey of 8,098 people.

 

A 2002 survey found that about 3.5% of all people in France were being prescribed antidepressants, compared to 1.7% in 1992, often for conditions other than depression and often not in line with authorizations or guidelines[32] Between 1996 and 2004 in British Columbia, antidepressant use increased from 3.4% to 7.2% of the population[33] Data from the Netherlands indicated an increasing rate of prescriptions of SSRIs, and an increasing duration of treatment.[34]

 

Surveys indicate that antidepressant use, particularly of SSRIs, has increased rapidly in most developed countries, driven by an increased awareness of depression together with the availability and commercial promotion of new antidepressants.[35] Antidepressants are also increasingly used worldwide for non-depressive patients as studies continue show the potential of immunomodulatory, analgesic and anti-inflammatory properties in antidepressants.

 

The choice of particular antidepressant is reported to be based, in the absence of research evidence of differences in efficacy, on seeking to avoid certain side effects, and taking into account comorbid (co-occurring) psychiatric disorders, specific clinical symptoms and prior treatment history[36]

 

It is also reported that, despite equivocal evidence of a significant difference in efficacy between older and newer antidepressants, clinicians perceive the newer drugs, including SSRIs and SNRIs, to be more effective than the older drugs (tricyclics and MAOIs).[37] A survey in the UK found that male general physicians were more likely to prescribe antidepressants than female doctors.[38]

 

Mechanisms of action

The therapeutic effects of antidepressants are believed to be related to their effects on neurotransmitters. Monoamine oxidase inhibitors (MAOIs) block the break-down of monoamine neurotransmitters (serotonin and norepinephrine) by inhibiting the enzymes which oxidize them, thus leaving higher levels still active in the brain (synaptic cleft).

 

Tricyclic antidepressants (TCAs) prevent the reuptake of various neurotransmitters, including serotonin, norepinephrine, and dopamine. Selective serotonin reuptake inhibitors (SSRIs) more specifically prevent the reuptake of serotonin (thereby increasing the level of active serotonin in synapses of the brain). Other novel antidepressants specifically affect serotonin and other neurotransmitters.

 

A theory centered on neurotransmitter effects appears to be incomplete, however. Neurotransmitter levels are altered as soon as the antidepressant chemicals build up in the bloodstream, but effects on mood appear to occur several days or weeks later.

 

One explanation of this holds that the "down-regulation" of neurotransmitter receptors—an apparent consequence of excess signaling and a process that takes several weeks—is actually the mechanism responsible for the alleviation of depressive symptoms. Another hypothesis is that antidepressants may have some longer-term effects due to the promotion of neurogenesis in the hippocampus, an effect found in mice[40][41] Other animal research suggests that antidepressants can also affect the expression of genes in brain cells, by influencing "clock genes".[42]

 

New research suggests that delayed onset of clinical effects from antidepressants indicates involvement of adaptive changes in antidepressant effects. Rodent studies have consistently shown upregulation of the 3, 5-cyclic adenosine monophosphate (cAMP) system induced by different types of chronic but not acute antidepressant treatment including serotonin and norepinephrine uptake inhibitors, monoamine oxidase inhibitors, tricyclic antidepressants, lithium and electroconvulsions. cAMP is synthesized from adenosine 5-triphosphate (ATP) by adenylyl cyclase and metabolized by cyclic nucleotide phosphodiesterases (PDEs).[43] Data also suggest antidepressants to have the ability of modulating neural plasticity in longterm administration.[44]

 

Therapeutic efficacy

There is a large amount of research evaluating the potential therapeutic effects of antidepressants, whether through efficacy studies under experimental conditions (including randomized clinical trials) or through studies of "real world" effectiveness. A sufficient response to a drug is often defined as at least a 50% reduction in self-reported or observed symptoms, with a partial response often defined as at least a 25% reduction. The term remission indicates a virtual elimination of depression symptoms, albeit with the risk of a recurrence of symptoms or complete relapse. Full remission or recovery signifies a full sustained return to a "normal" psychological state with full functioning.

 

Clinical guidelines

The American Psychiatric Association 2000 Practice Guideline for the Treatment of Patients with Major Depressive Disorder [15] indicates that, if preferred by the patient, antidepressant medications may be provided as an initial primary treatment for mild major depressive disorder; antidepressant medications should be provided for moderate to severe major depressive disorder unless electroconvulsive therapy is planned; and a combination of antipsychotic and antidepressant medications or electroconvulsive therapy should be used for psychotic depression. It states that efficacy is generally comparable between classes and within classes and that the initial selection will largely be based on the anticipated side effects for an individual patient, patient preference, quantity and quality of clinical trial data regarding the medication, and its cost.

 

The UK National Institute for Clinical Excellence (NICE) 2004 guidelines indicate that antidepressants should not be used for the initial treatment of mild depression, because the risk-benefit ratio is poor; that for moderate or severe depression an SSRI is more likely to be tolerated than a tricyclic; and that antidepressants for severe depression should be combined with a psychological treatment such as Cognitive Behavioural Therapy. [16]

 

Long-term use

The therapeutic effects of antidepressants typically do not continue once the course of medication ends, resulting in a high rate of relapse. A recent meta-analysis of 31 placebo-controlled antidepressant trials, mostly limited to studies covering a period of one year, found that 18% of patients who had responded to an antidepressant relapsed while still taking it, compared to 41% whose antidepressant was switched for a placebo.[75] The American Psychiatric Association guidelines advise four to five months of continuation treatment on an antidepressant following the resolution of symptoms. For patients with a history of depressive episodes, the British Association for Psychopharmacology's 2000 Guidelines for Treating Depressive Disorders with Antidepressants advise remaining on an antidepressant for at least six months and as long as five years or indefinitely.

 

Whether or not someone relapses after stopping an antidepressant does not appear to be related to the duration of prior treatment, however, and gradual loss of therapeutic benefit during the course also occurs. A strategy involving the use of pharmacotherapy in the treatment of the acute episode, followed by psychotherapy in its residual phase, has been suggested by some studies.[76][77]

 

Medication failure

Approximately 30% of patients have remission of depression with medications.[78] For patients with inadequate response, either adding sustained-release bupropion (initially 200 mg per day then increase by 100 mg up to total of 400 mg per day) or buspirone (up to 60 mg per day) for augmentation as a second drug can cause remission in approximately 30% of patients,[79] while switching medications can achieve remission in about 25% of patients.[80]

 

Tolerance and dependence

Most antidepressants, including the SSRIs and tricyclics, are known to produce tolerance (i.e. a decrease in the effects of a drug over time), and withdrawal (particularly if abrupt) may produce adverse effects, which can range from mild to extremely severe.

 

Antidepressants do not seem to have all of the same addictive qualities as other substances such as nicotine, caffeine, cocaine, or other stimulants - in other words, while antidepressants may cause dependence and withdrawal they do not seem to cause uncontrollable urges to increase the dose due to euphoria or pleasure, and thus do not meet the strict definition of an addictive substance. However, antidepressants do meet the World Health Organisation definition of "dependency-inducing", and indeed the SSRIs are listed by the organisation as among the most strongly dependency-inducing substances in existence.

 

If an SSRI medication is suddenly discontinued, it may produce both somatic and psychological withdrawal symptoms, a phenomenon known as "SSRI discontinuation syndrome" (Tamam & Ozpoyraz, 2002). When the decision is made to stop taking antidepressants it is common practice to "wean" off of them by slowly decreasing the dose over a period of several weeks or months, although often this will reduce the severity of the discontinuation reaction, rather than prevent it. Most cases of discontinuation syndrome last between one and four weeks, though there are examples of patients (especially those who have used the drugs for longer periods of time, or at a higher dose) experiencing adverse effects such as impaired concentration, poor short-term memory, elevated anxiety and sexual dysfunction, for months or even years after discontinuation.

 

It is generally not a good idea to take antidepressants without a prescription. The selection of an antidepressant and dosage suitable for a certain case and a certain person is a lengthy and complicated process, requiring the knowledge of a professional. Certain antidepressants can initially make depression worse, can induce anxiety, or can make a patient aggressive, dysphoric or acutely suicidal. In certain cases, an antidepressant can induce a switch from depression to mania or hypomania, can accelerate and shorten a manic cycle (i.e. promote a rapid-cycling pattern), or can induce the development of psychosis (or just the re-activation of latent psychosis) in a patient with depression who was not psychotic before the antidepressant.

 

Side effects

Antidepressants can often cause side effects, and an inability to tolerate these is the most common cause of discontinuing an otherwise working medication.

 

Side effects of SSRIs: Nausea, diarrhea, headaches. Sexual side effects are also common with SSRIs, such as loss of libido, failure to reach orgasm and erectile problems. Serotonin syndrome is also a worrying condition associated with the use of SSRIs. The Food and Drug Administration has included Black Box warnings on all SSRIs stating how they double suicidality (from 2 in 1,000 to 4 in 1,000) in children and adolescents who are prescribed these drugs.

 

Side effects of TCAs (tricyclic antidepressants): Fairly common side effects include dry mouth, blurred vision, drowsiness, dizziness, tremors, sexual problems, skin rash, and weight gain or loss.

 

Side effects of MAOIs (monoamine oxidase inhibitors): Rare side effects of MAOIs like phenelzine (brand name: Nardil) and tranylcypromine (brand name: Parnate) include liver inflammation, heart attack, stroke, and seizures.Serotonin syndrome is a side effect of MAOIs and SSRIs when they are combined.

 

Although recent drugs may have fewer side effects, patients sometimes report severe side effects associated with their discontinuation, particularly with paroxetine and venlafaxine. Additionally, a certain percentage of patients do not respond to antidepressant drugs. Another advantage of some newer antidepressants is they can show effects within as few as five days, whereas most take four to six weeks to show a change in mood. However, some studies show that these medications might be even more likely to result in moderate to severe sexual dysfunction. However, there are medications in trials that appear to show an improved profile in regard to sexual dysfunction and other key side effects.

 

MAO inhibitors can produce a potentially lethal hypertensive reaction if taken with foods that contain high levels of tyramine, such as mature cheese, cured meats or yeast extracts. Likewise, lethal reactions to both prescription and over the counter medications have occurred. Any patient currently undergoing therapy with an MAO inhibiting medication should be monitored closely by the prescribing physician and always consulted before taking an over the counter or prescribed medication. Such patients should also inform emergency room personnel and information should be kept with one's identification indicating the fact that the holder is on MAO inhibiting medications. Some doctors even suggest the use of a medical alert ID bracelet. Although the reactions in question are dramatic when they happen, the total number of deaths due to interactions and dietary concerns are comparable to over-the-counter medications.

 

Antidepressants should be used with great care, usually in conjunction with mood stabilisers, in the treatment of bipolar disorder, as they can exacerbate symptoms of mania. They have also been known to trigger mania or hypomania in some patients with bipolar disorder and in a small percentage of patients with depression.[17] SSRIs are the antidepressants most frequently associated with this side effect.

 

In particular, it has been noted that the most dangerous period for suicide in a patient with depression is immediately after treatment has commenced, as antidepressants may reduce the symptoms of depression such as psychomotor retardation or lack of motivation before mood starts to improve. Although this appears to be a paradox, studies indicate the suicidal ideation is a relatively common component of the initial phases of antidepressant therapy, and it may be even more prevalent in younger patients such as pre-adolescents and teenagers. It is strongly recommended that other family members and loved ones monitor the young patient's behavior, especially in the first eight weeks of therapy, for any signs of suicidal ideation or behaviors.

 

Until the black box warnings on these drugs were issued by FDA as well as by agencies in other nations, side effects and alerting families to risk were largely ignored and downplayed by manufacturers and practitioners. This may have resulted in some deaths by suicide although direct proof for such a link is largely anecdotal. The higher incidence of suicide ideation reported in a number of studies has drawn attention and caution in how these drugs are used.

 

People under the age of 24 who suffer from depression are warned that the use of antidepressants could increase the risk of suicidal thoughts and behaviour. Federal health officials unveiled Proposed changes to the labels on antidepressant drugs in December 2006 to warn people of the inherent danger.

 

On September 6, 2007, the Centers for Disease Control and Prevention reported suicide rate in American adolescents (especially girls, 10 to 24 years old) increased 8% (2003 to 2004), the largest jump in 15 years.[84] Specifically, in 2004 - 4,599 suicides in Americans ages 10 to 24, up from 4,232 in 2003, for a rate of 7.32 per 100,000 people that age. Before, the rate dropped to 6.78 per 100,000 in 2003 from 9.48 per 100,000 in 1990. The findings also reinforced the fact that antidepressant drugs reduce suicide risk. Psychiatrists found that the increase is due to the decline in prescriptions of antidepressant drugs like Prozac to young people since 2003, leaving more cases of serious depression untreated. In a December 2006 study, The American Journal of Psychiatry said that a decrease in antidepressant prescriptions to minors of just a few percentage points coincided with a 14 percent increase in suicides in the United States; in the Netherlands, the suicide rate was 50% up, upon prescription drop.[85] The critics of this study contend that the US "2004 suicide figures were compared simplistically with the previous year, rather than examining the change in trends over several years".[86] The pitfalls of such attempts to infer a trend using just two data points (years 2003 and 2004) are further demonstrated by the fact that, according to the new epidemiological data, the suicide rate in 2005 in children and adolescents actually declined despite the continuing decrease of SSRI prescriptions. "It is risky to draw conclusions from limited ecologic analyses of isolated year-to-year fluctuations in antidepressant prescriptions and suicides. One promising epidemiological approach involves examining the associations between trends in psychotropic medication use and suicide over time across a large number of small geographic regions. Until the results of more detailed analyses are known, prudence dictates deferring judgment concerning the public health effects of the FDA warnings."[87][88] Subsequest follow-up studies have supported the hypothesis that antidepressant drugs reduce suicide risk.[89] [90] However, the conclusion that societal suicide rate decreases are due to antidepressant prescription is extraordinarily dubious given the plethora of confounding variables.

 

Sexual dysfunction

Sexual dysfunction is a very common side effect, especially with SSRIs. Common sexual side effects include problems with libido (sexual desire), lack of interest in sex, and anorgasmia (trouble achieving orgasm). [18] Although usually reversible, these sexual side effects can, in rare cases, last for months or years after the drug has been completely withdrawn. This disorder is known as Post SSRI Sexual Dysfunction.

 

Bupropion, a dual reuptake inhibitor (NE and DA), in many cases results in a moderately increased libido, due to increased dopamine activity. This effect is also seen with dopamine reuptake inhibitors, CNS stimulants and dopamine agonists, and is due to increases in testosterone production (due to inhibition of prolactin) and increased nitric oxide synthesis. Apomorphine, nefazodone and nitroglycerin have been shown to reverse some sexual dysfunction via increased nitric oxide activity. MAOIs are reported to have fewer negative effects on sexual function and libido, particularly moclobemide at a 1.9% rate of occurrence. Betanechol has been reported to reverse MAOI-induced sexual dysfunction via its cholinergic agonist properties (Gross 1982).

 

In order for the physician to select the appropriate response, the patient should provide the physician with information to distinguish between reduced libido (little or no desire for sex), reduced sexual function (impotence, vaginal dryness) and anorgasmia, as these have separate causes and prompt different treatment.

 

Thymoanesthesia

Closely related to sexual side effects is the phenomenon of emotional blunting, or mood anesthesia. Many users of SSRIs complain of apathy, lack of motivation, emotional numbness, feelings of detachment, and indifference to surroundings. They may describe this as a feeling of "not caring about anything anymore." All SSRIs, SNRIs, and serotonergic TCAs are liable to cause this effect to varying degrees, especially at higher dosages.

 

REM sleep

It is well recognized that virtually all major antidepressant drugs but trimipramine suppress REM sleep and it has, in fact, been proposed that the clinical efficacy of these drugs largely derives from their suppressant effects on REM sleep. The three major classes of antidepressant drugs, monoamine oxidase inhibitors (MAOIs), tricyclic antidepressants (TCAs) and selective serotonin reuptake inhibitors (SSRIs), profoundly suppress REM sleep.[19] The MAOIs virtually completely abolish REM sleep, while the TCAs and SSRIs have been shown to produce immediate (40-85%) and sustained (30-50%) reductions in REM sleep. Abrupt discontinuation of MAOIs can cause a temporary phenomenon known as "REM rebound" in which the patient experiences extremely vivid dreams and nightmares.

 

Weight gain

Many antidepressants in all categories are associated with weight gain usually in the range of 10-50 pounds but not uncommonly upwards of 100 pounds. The specific cause is unknown, but it is known that antidepressants are associated with increased cravings (usually for high fat carbohydrates), an inability to feel full despite ingestion of adequate calories, low energy levels and increased daytime sleepiness which can lead to overeating and a lack of desire to exercise, and dry mouth which can lead to ingestion of calorie-laden beverages. Eating low fat, low protein carbohydrate snacks and carbohydrate-rich dinners allows the brain to make serotonin which then controls appetite and balances mood. Carbohydrates thus eaten, as part of a balanced diet, by virtue of their effect on brain serotonin levels, thus support weight loss in the setting of antidepressant weight gain.[91][92]

 

Controversy

Several studies have stimulated doubt about the effectiveness of antidepressants. A 2002 study cited that the difference between antidepressants and placebo is close to negligible.[93]

 

The paper in question has been severely criticized by independent researchers, however. One reason for this is that it deals almost exclusively with the SSRI class of medication. In leveling criticism against the efficacy of SSRIs, critics state, it is not the best paper, merely the most widely known one. Also, other classes of antidepressants have demonstrated superior efficacy, and it has been argued that this paper is "throwing the baby out with the bathwater", while its thrust should in fact be leveled at the serotonin hypothesis of depression.

 

Furthermore, not all patients necessarily respond to a given medication, studies do not always address dosage versus drug-placebo differences for those who do. Data submitted to the FDA can also underestimate how a drug will perform in clinic practice, as studies sometimes are designed as much for marketing purposes as they are to estimate the magnitude of a medication's effects.[94]

 

Through a Freedom of Information Act request, two psychologists obtained 47 studies used by the FDA for approval of the six antidepressants prescribed most widely between 1987-99. Overall, antidepressant pills worked 18% better than placebos, a statistically significant difference, "but not meaningful for people in clinical settings", says University of Connecticut psychologist Irving Kirsch. He and co-author Thomas Moore released their findings in "Prevention and Treatment", an e-journal of the American Psychological Association.[20]

 

More than half of the 47 studies found that patients on antidepressants improved no more than those on placebos, Kirsch says. "They should have told the American public about this. The drugs have been touted as much more effective than they are." He says studies finding no benefit have been mentioned only on labeling for Celexa, the most recently approved drug. The others included in his evaluation: Prozac, Paxil, Zoloft, Effexor and Serzone.

 

Dr Joseph Glenmullen, a Harvard psychiatrist, has written a book on the subject for the layperson; see link below.

 

In 2007, anti-depressants became the most prescribed drug in the United States, causing more debate over the issue. Some doctors believe this is a positive sign that people are finally seeking help for their issues. Others disagree, saying that this shows that people are becoming too dependent on anti-depressants. [21]

 

References

1. ^ Vega, JA., Mortimer, AM., Tyson, PJ. (2003) Conventional antipsychotic prescription in unipolar depression, I: An audit and recommendations for practice The Journal of clinical psychiatry 64(5), pp. 568-574.
2. ^ Petty F, Trivedi MH, Fulton M, Rush AJ. (1995) Benzodiazepines as antidepressants: does GABA play a role in depression? Biological Psychiatry. 38(9):578-91
3. ^ Weber, M.M. & Emrich, H.M. (1988) Current and historical concepts of opiate treatment in psychiatric disorders. International Journal of Clinical Psychopharmacology. Jul;3(3):255-66.
4. ^ Czygan, F.C. (2003) From a 2500 year old apotropic comes a current antidepressive. The cultural history and mistique of St. John's wort Pharm Unserer Zeit. 32(3):184-90
5. ^ SELIKOFF IJ, ROBITZEK EH (1952). "Tuberculosis chemotherapy with hydrazine derivatives of isonicotinic acid". Dis Chest 21 (4): 385–438. PMID 14906149.
6. ^ a b c Weissman, Myrna M. (2001). Treatment of depression: bridging the 21st century. Washington, D.C: American Psychopathological Association, 10-11. ISBN 0-88048-397-0.
7. ^ Healy, David (1996). The Psychopharmacologists: Interviews. London: Chapman & Hall, 8. ISBN 1-86036-008-4.
8. ^ Healy, David (1998). The Psychopharmacologists: Volume 2. A Hodder Arnold Publication, 132-134. ISBN 1-86036-010-6.
9. ^ ROBITZEK EH, SELIKOFF IJ, MAMLOK E, TENDLAU A (1953). "Isoniazid and its isopropyl derivative in the therapy of tuberculosis in humans: comparative therapeutic and toxicologic properties". Dis Chest 23 (1): 1–15. PMID 12998444.
10. ^ a b c d López-Muñoz F, Alamo C, Juckel G, Assion HJ (2007). "Half a century of antidepressant drugs: on the clinical introduction of monoamine oxidase inhibitors, tricyclics, and tetracyclics. Part I: monoamine oxidase inhibitors". J Clin Psychopharmacol 27 (6): 555–9. doi:10.1097/jcp.0b013e3181bb617. PMID 18004120.
11. ^ Time Magazine (1957) "Psychic Energizer"
12. ^ Kuhn, R. (1958) The Treatment of Depressive States with G 22355 (Imipramine Hydrochloride). American Journal of Psychiatry, 115:459-464, November
13. ^ Biel JH. (1964). Some rationales for the development of antidepressant drugs. Molecular modification in drug design. Adv Chem; 45: 114–39
14. ^ Pletscher, A. (1991) The discovery of antidepressants: A winding path. Journal of Cellular and Molecular Life Sciences, Volume 47(1)
15. ^ Healy, D.M.D. (1999)
16. ^ The Three Faces of the Antidepressants: A Critical Commentary on the Clinical-Economic Context of Diagnosis. Journal of Nervous & Mental Disease. 187(3):174-180
17. ^ Domino, E.F. (1999) [1] History of Modern Psychopharmacology: A Personal View With an Emphasis on Antidepressants Psychopharmacology and Psychosomatic Research, Special Issue
18. ^ Wong DT, Bymaster FP, Horng JS, Molloy BB. (1975) [2] A new selective inhibitor for uptake of serotonin into synaptosomes of rat brain: 3-(p-trifluoromethylphenoxy) N-methyl-3-phenylpropylamine]. Journal of Pharmacology and Experimental Therapeutics. 193, p 804–811
19. ^ Linde K, Ramirez G, Mulrow CD, Pauls A, Weidenhammer W & Melchart D (1996) St John’s wort for depression – an overview and meta-analysis of randomised clinical trials. British Medical Journal 313, p253–258.
20. ^ Muller, W.E. (2003) Current St John's wort research from mode of action to clinical efficacy. Pharmacol Res. Feb;47(2):101-9
21. ^ Nathan, P.J. (2001) J Hypericum perforatum (St John's Wort): a non-selective reuptake inhibitor? A review of the recent advances in its pharmacology. Psychopharmacol. 2001 Mar;15(1):47-54.
22. ^ Freeman, H. (1996) Tolerability and safety of novel antidepressants. European Psychiatry, 11(supplement 4), p206.
23. ^ Lacasse J, Leo J (2005). "Serotonin and depression: a disconnect between the advertisements and the scientific literature". PLoS Med 2 (12): e392. PMID 16268734. Full text
24. ^ Uz T, Ahmed R, Akhisaroglu M, Kurtuncu M, Imbesi M, Dirim Arslan A, Manev H (2005). "Effect of fluoxetine and cocaine on the expression of clock genes in the mouse hippocampus and striatum". Neuroscience 134 (4): 1309-16. PMID 15994025.
25. ^ Yuferov V, Butelman E, Kreek M (2005). "Biological clock: biological clocks may modulate drug addiction". Eur J Hum Genet 13 (10): 1101-3. PMID 16094306.
26. ^ Manev H, Uz T (2006). "Clock genes as a link between addiction and obesity". Eur J Hum Genet 14 (1): 5. PMID 16288309.Full text
27. ^ www.psychiatrist.com/pcc/pccpdf/v06n04/v06n0403.pdf (PDF).
28. ^ (National Institute for Clinical Excellence, 2004)
29. ^ [3]Stagnitti,M. (2005) Antidepressant Use in the US Civilian Non-Insitutionalised Population, 2002. Statistical Brief #77. Rockville,MD: Medical Expenditure Panel, Agency for Healthcare Research and Quality.
30. ^ Sleath, B. & Shih, YC. (2003) [4] Sociological influences on antidepressant prescribing. Soc Sci Med. Mar;56(6):1335-44.
31. ^ [5] Depression could be overdiagnosed: Up to 25% may be reacting normally to stresses, study says
32. ^ Olie JP, Elomari F, Spadone C, Lepine JP. (2002) Antidepressant consumption in the global population in France Encephale. Sep-Oct;28(5 Pt 1):411-7.
33. ^ Raymond CB, Morgan SG, Caetano PA. (2007) Antidepressant utilization in british columbia from 1996 to 2004: increasing prevalence but not incidence. Psychiatr Serv. 2007 Jan;58(1):79-84.
34. ^ Meijer W, Heerdink E, Leufkens H, Herings R, Egberts A, Nolen W (2004). "Incidence and determinants of long-term use of antidepressants". Eur J Clin Pharmacol 60 (1): 57-61. PMID 14985889.
35. ^ McManus P, Mant A, Mitchell PB, Montgomery WS, Marley J, Auland ME. (2000) Recent trends in the use of antidepressant drugs in Australia, 1990-1998.Med J Aust. November 6;173(9):458-61.
36. ^ Mark Zimmerman, Michael Posternak, Michael Friedman, Naureen Attiullah, Scott Baymiller, Robert Boland, Stacie Berlowitz, Shahzad Rahman, Kirsten Uy, Steve Singer (2004) Which Factors Influence Psychiatrists’ Selection of Antidepressants? Am J Psychiatry 161:1285-1289
37. ^ Petersen T, Dording C, Neault NB, Kornbluh R, Alpert JE, Nierenberg AA, Rosenbaum JF, Fava M. (2002) A survey of prescribing practices in the treatment of depression. Prog Neuropsychopharmacol Biol Psychiatry. Jan;26(1):177-87.
38. ^ BBC News (July 2002) Male GPs depression pills 'bias'
39. ^ The number of prescriptions was calculated as the total of prescriptions for the corresponding generic and brand-name drugs using data from the charts for generic and brand-name drugs.Top 200 generic drugs by units in 2006. Top 200 brand-name drugs by units.. Drug Topics, Mar 5, 2007. Retrieved on 2007-12-24.
40. ^ [6] Malberg JE, Eisch AJ, Nestler EJ, Duman RS. Chronic antidepressant treatment increases neurogenesis in adult rat hippocampus. J Neurosci. 2000 December 15;20(24):9104-10.
41. ^ [7]Manev H, Uz T, Smalheiser NR, Manev R. Antidepressants alter cell proliferation in the adult brain in vivo and in neural cultures in vitro. Eur J Pharmacol. 2001 January 5;411(1-2):67-70.
42. ^ [8] Uz T, Ahmed R, Akhisaroglu M, Kurtuncu M, Imbesi M, Dirim Arslan A, Manev H. Effect of fluoxetine and cocaine on the expression of clock genes in the mouse hippocampus and striatum. Neuroscience. 2005;134(4):1309-16.
43. ^ Zhang HT, Huang Y, Mishler K, Roerig SC, O'Donnell JM. Interaction between the antidepressant-like behavioral effects of beta adrenergic agonists and the cyclic AMP PDE inhibitor rolipram in rats. Psychopharmacology (Berl). 2005 Oct;182(1):104-15
44. ^ Carboni L, Vighini M, Piubelli C, Castelletti L, Milli A, Domenici E. Proteomic analysis of rat hippocampus and frontal cortex after chronic treatment with fluoxetine or putative novel antidepressants: CRF1 and NK1 receptor antagonists. Eur Neuropsychopharmacol. 2006 Oct;16(7):521-37.
45. ^ O'Brien SM, Scully P, Scott LV, Dinan TG. "Cytokine profiles in bipolar affective disorder: focus on acutely ill patients." J Affect Disord. 2006 Feb;90(2-3):263-7.
46. ^ Obuchowicz E, Marcinowska A, Herman ZS. "Antidepressants and cytokines--clinical and experimental studies" Psychiatr Pol. 2005 Sep-Oct;39(5):921-36.
47. ^ Hong CJ, Yu YW, Chen TJ, Tsai SJ."Interleukin-6 genetic polymorphism and Chinese major depression". Neuropsychobiology. 2005;52(4):202-5.
48. ^ Elenkov IJ, Iezzoni DG, Daly A, Harris AG, Chrousos GP. "Abstract Cytokine dysregulation, inflammation and well-being". Neuroimmunomodulation. 2005;12(5):255-69
49. ^ Kubera M, Maes M, Kenis G, Kim YK, Lason W. "Effects of serotonin and serotonergic agonists and antagonists on the production of tumor necrosis factor alpha and interleukin-6" Psychiatry Res. 2005 April 30;134(3):251-8.
50. ^ Elenkov IJ, Iezzoni DG, Daly A, Harris AG, Chrousos GP. "Cytokine dysregulation, inflammation and well-being". Neuroimmunomodulation. 2005;12(5):255-69
51. ^ Diamond M, Kelly JP, Connor TJ. "Antidepressants suppress production of the Th1 cytokine interferon-gamma, independent of monoamine transporter blockade". Eur Neuropsychopharmacol. 2006 Oct;16(7):481-90.
52. ^ Kubera M, Lin AH, Kenis G, Bosmans E, van Bockstaele D, Maes M. "Anti-Inflammatory effects of antidepressants through suppression of the interferon-gamma/interleukin-10 production ratio." J Clin Psychopharmacol. 2001 Apr;21(2):199-206
53. ^ Maes M."The immunoregulatory effects of antidepressants". Hum Psychopharmacol. 2001 Jan;16(1):95-103
54. ^ Maes M, Kenis G, Kubera M, De Baets M, Steinbusch H, Bosmans E."The negative immunoregulatory effects of fluoxetine in relation to the cAMP-dependent PKA pathway". Int Immunopharmacol. 2005 Mar;5(3):609-18.
55. ^ Brustolim D, Ribeiro-dos-Santos R, Kast RE, Altschuler EL, Soares MB. "A new chapter opens in anti-inflammatory treatments: the antidepressant bupropion lowers production of tumor necrosis factor-alpha and interferon-gamma in mice." Int Immunopharmacol. 2006 Jun;6(6):903-7
56. ^ Moulin DE, Clark AJ, Gilron I, Ware MA, Watson CP, Sessle BJ, Coderre T, Morley-Forster PK, Stinson J, Boulanger A, Peng P, Finley GA, Taenzer P, Squire P, Dion D, Cholkan A, Gilani A, Gordon A, Henry J, Jovey R, Lynch M, Mailis-Gagnon A, Panju A, Rollman GB, Velly A; Canadian Pain Society.Pharmacological management of chronic neuropathic pain - consensus statement and guidelines from the Canadian Pain Society. Pain Res Manag. 2007 Spring;12(1):13-21
57. ^ Jones CK, Eastwood BJ, Need AB, Shannon HE. Analgesic effects of serotonergic, noradrenergic or dual reuptake inhibitors in the carrageenan test in rats: evidence for synergism between serotonergic and noradrenergic reuptake inhibition.Neuropharmacology. 2006 Dec;51(7-8):1172-80.
58. ^ Kulmatycki KM, Jamali F. "Drug disease interactions: role of inflammatory mediators in depression and variability in antidepressant drug response". J Pharm Pharm Sci. 2006;9(3):292-306.
59. ^ O'Brien SM, Scott LV, Dinan TG. "Cytokines: abnormalities in major depression and implications for pharmacological treatment". Hum Psychopharmacol. 2004 Aug;19(6):397-403.
60. ^ Anderson IM (2000) Selective serotonin reuptake inhibitors versus tricyclic antidepressants: a meta-analysis of efficacy and tolerability Journal of Affective Disorders. 58(1), 19-3
61. ^ MacGillivray, S., Arroll,B., Hatcher,S., Ogston,S., Reid,I., Sullivan, F., Williams,B., Crombie,I. (2003) Efficacy and tolerability of selective serotonin reuptake inhibitors compared with tricyclic antidepressants in depression treated in primary care: systematic review and meta-analysis BMJ. May 10; 326(7397): 1014.
62. ^ Parker G, Roy K, Wilhelm K, Mitchell P. (2001) Assessing the comparative effectiveness of antidepressant therapies: a prospective clinical practice study. J Clin Psychiatry. Feb;62(2):117-25.
63. ^ Moncrieff J, Wessely S, Hardy R. (2004) Active placebos versus antidepressants for depression. Cochrane Database Syst Rev. (1):CD003012.
64. ^ Lotufo-Neto, F., Trivedi, M., & Thase, M.E. (1999) Meta-Analysis of the Reversible Inhibitors of Monoamine Oxidase Type A Moclobemide and Brofaromine for the Treatment of Depression Neuropsychopharmacology 20 226-247.10.1038
65. ^ Linde K, Mulrow CD, Berner M, Egger M. (2005) St John's wort for depression. Cochrane Database Syst Rev. 2005 April 18;(2):CD000448.
66. ^ Jureidini JN, Doecke CJ, Mansfield PR, Haby MM, Menkes DB, Tonkin AL. (2004) Efficacy and safety of antidepressants for children and adolescents. BMJ. 2004 April 10;328(7444):879-83.
67. ^ Lakhan SE; Hagger-Johnson G. The impact of prescribed psychotropics on youth. Clinical Practice and Epidemiology in Mental Health 2007;3(21).
68. ^ Baghai, TC., Moller, HJ, Rupprecht, R. (2006) Recent progress in pharmacological and non-pharmacological treatment options of major depression. Curr Pharm Des. ;12(4):503-15.
69. ^ a b Ruhe HG, Huyser J, Swinkels JA, Schene AH. (2006)Switching antidepressants after a first selective serotonin reuptake inhibitor in major depressive disorder: a systematic review. J Clin Psychiatry. Dec;67(12):1836-55.
70. ^ Tranter, R., O'Donovan, CO., Chandarana, P., Kennedy, S. (2002) Prevalence and outcome of partial remission in depression J Psychiatry Neurosci. July; 27(4): 241–247.
71. ^ Byrne, SE. & Rothschild, AJ. (1999) Loss of antidepressant efficacy during maintenance therapy: possible mechanisms and treatments. J Clin Psychiatry. Jun;59(6):279-88.
72. ^ Mischoulon D, Nierenberg AA, Kizilbash L, Rosenbaum JF, Fava M. (2000) Strategies for managing depression refractory to selective serotonin reuptake inhibitor treatment: a survey of clinicians. Can J Psychiatry. Jun;45(5):476-81.
73. ^ DeBattista C, Lembke A. (2005) Update on augmentation of antidepressant response in resistant depression. Curr Psychiatry Rep. Dec;7(6):435-40.
74. ^ Lam RW, Wan DD, Cohen NL, Kennedy SH. (2002) Combining antidepressants for treatment-resistant depression: a review. J Clin Psychiatry. Aug;63(8):685-93.
75. ^ Geddes JR et al. (2003) Relapse prevention with antidepressant drug treatment in depressive disorders: a systematic review. Lancet. February 22;361(9358):653-61.
76. ^ Fava GA, Park SK, Sonino N. (2006) Treatment of recurrent depression. Expert Rev Neurother. Nov;6(11):1735-40.
77. ^ Peterson, TJ. (2006) Enhancing the efficacy of antidepressants with psychotherapy Journal of Psychopharmacology, Vol. 20, No. 3 suppl, 19-28, 2006
78. ^ Trivedi MH, Rush AJ, Wisniewski SR, et al (2006). "Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: implications for clinical practice". The American journal of psychiatry 163 (1): 28-40. doi:10.1176/appi.ajp.163.1.28. PMID 16390886.
79. ^ Trivedi MH, Fava M, Wisniewski SR, et al (2006). "Medication augmentation after the failure of SSRIs for depression". N. Engl. J. Med. 354 (12): 1243-52. doi:10.1056/NEJMoa052964. PMID 16554526.
80. ^ Rush AJ, Trivedi MH, Wisniewski SR, et al (2006). "Bupropion-SR, sertraline, or venlafaxine-XR after failure of SSRIs for depression". N. Engl. J. Med. 354 (12): 1231-42. doi:10.1056/NEJMoa052963. PMID 16554525.
81. ^ Cohen, MD, Lee S.; Lori L. Altshuler, MD; Bernard L. Harlow, PhD; Ruta Nonacs, MD, PhD; D. Jeffrey Newport, MD; Adele C. Viguera, MD; Rita Suri, MD; Vivien K. Burt, MD, PhD; Victoria Hendrick, MD; Alison M. Reminick, BA; Ada Loughead, BA; Allison F. Vitonis, BA; Zachary N. Stowe, MD (February 1, 2006). "Relapse of Major Depression During Pregnancy in Women Who Maintain or Discontinue Antidepressant Treatment". Journal of the American Medical Association 295 (5): 499-507. American Medical Association. Retrieved on 2007-06-14.
82. ^ (July 12, 2006) "Relapse of Major Depression During Pregnancy in Women Who Maintain or Discontinue Antidepressant Treatment—Correction". JAMA 296 (2): 170. Retrieved on 2007-06-14.
83. ^ David Armstrong, "Drug Interactions: Financial Ties to Industry Cloud Major Depression Study At Issue: Whether It's Safe For Pregnant Women To Stay on Medication - JAMA Asks Authors to Explain". Wall Street Journal. July 11, 2006 (copy published on post-gazette.com)
84. ^ Suicide Rises in Youth; Antidepressant Debate Looms, Benedict Carey, New York Times, September 7, 2007
85. ^ Simon GE, Savarino J, Operskalski B, Wang PS (2006). "Suicide risk during antidepressant treatment". Am J Psychiatry 163 (1): 41–7. doi:10.1176/appi.ajp.163.1.41. PMID 16390887.
86. ^ Jureidini J (2007). "The black box warning: decreased prescriptions and increased youth suicide?". Am J Psychiatry 164 (12): 1907. doi:10.1176/appi.ajp.2007.07091463. PMID 18056248.
87. ^ Olfson M, Shaffer D (2007). "SSRI Prescriptions and the Rate of Suicide". Am J Psychiatry 164 (12): 1907–1908. doi:10.1176/appi.ajp.2007.07091467. PMID 18056247.
88. ^ Kung HC, Hoyert DL, Xu J, Murphy SL. N C H S - Health E Stats - Deaths: Preliminary Data for 2005 (htm). National Center for Health Statistics. Retrieved on 2007-12-12.
89. ^ Bridge JA, Iyengar S, Salary CB, et al (2007). "Clinical response and risk for reported suicidal ideation and suicide attempts in pediatric antidepressant treatment: a meta-analysis of randomized controlled trials". JAMA 297 (15): 1683–96. doi:10.1001/jama.297.15.1683. PMID 17440145.
90. ^ Beasley CM, Ball SG, Nilsson ME, et al (2007). "Fluoxetine and Adult Suicidality Revisited: An Updated Meta-Analysis Using Expanded Data Sources From Placebo-Controlled Trials". J Clin Psychopharmacol 27 (6): 682–686. doi:10.1097/jcp.0b013e31815abf21. PMID 18004137.
91. ^ Wurtman, J., Wurtman R., McDermott J., Levendusky P and Duca K. (2002). The Effect of a Novel Dietary Intervention on Weight Loss in Psychotropic Drug-Induced Obesity. Psychopharmacology Bulletin Vol. 36 No. 3 p. 55-59
92. ^ Wurtman, J. Marquis, N (2007) The Serotonin Power Diet, Rodale Books.
93. ^ [9]Kirsch I, Moore TJ, Scoboria A, Nicholls SS (2002a), The emperor's new drugs: an analysis of antidepressant medication data submitted to the U.S. Food and Drug Administration. Prevention & Treatment 5:Article 23
94. ^ Hollon, Steven D.; DeRubeis, Robert J.; Shelton, Richard C.; Weiss, Bahr The emperor's new drugs: Effect size and moderation effects. Prevention & Treatment. 5(1), July 2002, No Pagination Specified.
95. ^ [10] from Richard Zitrin & Carol M. Langford. "Hide and Secrets in Louisville" from "The Moral Compass of the American Lawyer". Ballantine Books, 1999
96. ^ [11] October 1998 GSK Internal Memo: Strategy of Concealment
97. ^ [12] Newstarget September 6, 2005
98. ^ World Health Organisation, Acupuncture: Review and Analysis of Reports on Controlled Clinical Trials, 2002
99. ^ ABC Australia Science News on acupuncuture.
100. ^ Hypericum Depression Trial Study Group (2002). Effect of Hypericum perforatum (St John's Wort) in Major Depressive Disorder: A Randomized Controlled Clinical Trial. JAMA, 287 (14):1807-1814
101. ^ Roberto Delle Chiaie, Paolo Pancheri and Pierluigi Scapicchio. (2002). Efficacy and tolerability of oral and intramuscular S-adenosyl- L-methionine 1,4-butanedisulfonate (SAMe) in the treatment of major depression: comparison with imipramine in 2 multicenter studies. Am J Clin Nutr, 76 (5): 1172S-1176S
102. ^ Mischoulon D, Fava M. (2002). Role of S-adenosyl-L-methionine in the treatment of depression: a review of the evidence. Am J Clin Nutr, 76 (5): 1158S-61S.
103. ^ "Drug 'treats depression in hours'", BBC, 2006-08-07. Retrieved on 2006-09-17.

 

[edit] Additional reading

 

Closing button for this Autism and Asperger's syndrome fact sheet

Click here for the full range of Asperger's and Autism fact sheets at www.autism-help.org
This autism fact sheet is licensed under the GNU Free Documentation. It is derivative of an Autism and Asperger's syndrome-related articles at http://en.wikipedia.org

     
   
Medication can be useful for autistic children in some cases of behaviors, seizures and treating comorbid disorders associated with Autism