Serotonin–norepinephrine reuptake inhibitor

Source: Wikipedia, the free encyclopedia.
(Redirected from
Serotonin-norepinephrine reuptake inhibitors
)
Serotonin–norepinephrine reuptake inhibitor
Drug class
Duloxetine, an example of an SNRI.
Class identifiers
SynonymsSelective Serotonin–noradrenaline reuptake inhibitor; SNaRI
UseDepression; Anxiety; Pain; Obesity; Menopausal symptoms
Biological targetSerotonin transporter; Norepinephrine transporter
External links
MeSHD000068760
Legal status
In Wikidata

Serotonin–norepinephrine reuptake inhibitors (SNRIs) are a class of

attention-deficit hyperactivity disorder (ADHD), obsessive–compulsive disorder (OCD), and migraine prevention.[1] SNRIs are monoamine reuptake inhibitors; specifically, they inhibit the reuptake of serotonin and norepinephrine. These neurotransmitters are thought to play an important role in mood regulation. SNRIs can be contrasted with the selective serotonin reuptake inhibitors (SSRIs) and norepinephrine reuptake inhibitors
(NRIs), which act upon single neurotransmitters.

The human

noradrenaline transporter (NAT) are membrane transport proteins that are responsible for the reuptake of serotonin and noradrenaline from the synaptic cleft back into the presynaptic nerve terminal. Dual inhibition of serotonin and noradrenaline reuptake can offer advantages over other antidepressant drugs by treating a wider range of symptoms.[2] They can be especially useful in concomitant chronic or neuropathic pain.[3]

SNRIs, along with SSRIs and NRIs, are

tricyclic antidepressants (TCAs) and monoamine oxidase inhibitors (MAOIs), as the drugs of choice for the treatment of MDD due to their improved tolerability and safety profile.[4]

Medications

Timeline-SNRIs-2010
Timeline of approved SNRIs.

There are eight FDA approved SNRIs in the United States, with venlafaxine being the first drug to be developed in 1993 and levomilnacipran being the latest drug to be developed in 2013. The drugs vary by their other medical uses, chemical structure, adverse effects, and efficacy.[5]

Medication Brand name FDA Indications Approval Year Chemical structure Notes
Desvenlafaxine[6] Pristiq

Khedezla (ER)

  • Major depressive disorder[7]
2007
The active metabolite of venlafaxine. It is believed to work in a similar manner, though some evidence suggests lower response rates compared to venlafaxine and duloxetine. It was introduced by Wyeth in May 2008 and was then the third approved SNRI.[7]
Duloxetine[8] Cymbalta

Irenka

2004
Duloxetine
Duloxetine
Approved for the treatment of depression and neuropathic pain in August 2004. Duloxetine is contraindicated in patients with heavy alcohol use or chronic liver disease, as duloxetine can increase the levels of certain liver enzymes that can lead to acute hepatitis or other diseases in certain at risk patients. The risk of liver damage appears to be only for patients already at risk, unlike the antidepressant nefazodone, which, though rare, can spontaneously cause liver failure in healthy patients.[12] Duloxetine is also approved for major depressive disorder (MDD), generalized anxiety disorder (GAD), diabetic neuropathy, chronic musculoskeletal pain, including chronic osteoarthritis pain and chronic low back pain.[10] Duloxetine also undergoes hepatic metabolism and has been shown to cause inhibition of the hepatic cytochrome P450 enzyme CYP 2D6.[13] Caution should be taken when taking Duloxetine with other medications that are metabolized by CYP 2D6 as this may precipitate a potential drug-drug interaction.[13]
Levomilnacipran Fetzima
  • Major depressive disorder
2013
Levomilnacipran
Levomilnacipran
The
levorotating
isomer of milnacipran. Under development for the treatment of depression in the United States and Canada, it was approved by the FDA for treatment of MDD in July 2013.
Milnacipran Ixel
Savella
Impulsor
  • Fibromyalgia
  • Major depressive disorder[14]
1996
Milnacipran
Milnacipran
Shown to be significantly effective in the treatment of depression and fibromyalgia.[14] The Food and Drug Administration (FDA) approved milnacipran for treatment of fibromyalgia in the United States in January 2009, however it is not approved for depression in that country.[citation needed] Milnacipran has been commercially available in Europe and Asia for several years.[citation needed] It was first introduced in France in 1996.[citation needed]
Sibutramine Meridia 1997
An SNRI, which, instead of being developed for the treatment of depression, was widely marketed as an
appetite suppressant for weight loss purposes. Sibutramine was the first drug for the treatment of obesity to be approved in 30 years.[16] It has been associated with increased cardiovascular events and strokes and has been withdrawn from the market in several countries and regions including the United States in 2010.[17]
Tramadol Ultram
  • Acute and chronic pain
1977
A dual weak opioid and SNRI. It was approved by the FDA in 1995, though it has been marketed in Germany since 1977. The drug is used to treat acute and chronic pain. It has shown effectiveness in the treatment of fibromyalgia, though it is not specifically approved for this purpose. The drug is also under investigation as an antidepressant and for the treatment of neuropathic pain. It is related in chemical structure to venlafaxine. Due to being an opioid, there is risk of abuse and addiction, but it does have less abuse potential, respiratory depression, and constipation compared to other opioids (hydrocodone, oxycodone, etc.).[18]
Venlafaxine Effexor
  • Major depressive disorder
  • Generalized anxiety disorder
  • Social anxiety disorder
  • Panic disorder
  • Chronic pain syndromes[19]
  • Vasomotor symptoms of menopause (hot flashes)[20]
1994
The first and most commonly used SNRI. It was introduced by
vasomotor symptoms (hot flashes and night sweats) of menopause and may be as effective as hormone replacement therapy (HRT).[20]

History

In 1952,

side effects and significant safety and toxicity issues.[22]

Throughout the 1960s and 1970s, the catecholamine hypothesis of emotion and its relation to depression was of wide interest and that the decreased levels of certain neurotransmitters, such as norepinephrine, serotonin, and dopamine might play a role in the pathogenesis of depression. This led to the development of fluoxetine, the first SSRI. The improved safety and tolerability profile of the SSRIs in patients with MDD, compared with TCAs and MAOIs, represented yet another important advance in the treatment of depression.[22]

Since the late 1980s, SSRIs have dominated the antidepressant drug market. Today, there is increased interest in antidepressant drugs with broader

phenylethylamines. These substances are unrelated to TCA and other SSRIs. Venlafaxine blocks the neuronal reuptake of serotonin, noradrenaline, and, to a lesser extent, dopamine in the central nervous system. In contrast with several other antidepressant drugs, venlafaxine can induce a rapid onset of action mainly due to a subsequent norepinephrine reuptake inhibition.[23]
See timeline in figure 1.

Timeline
Timeline of development of antidepressant agents.

Mechanism of action

Monoamines are connected to the pathophysiology of depression. Symptoms may occur because concentrations of neurotransmitters, such as norepinephrine and serotonin, are insufficient, leading to downstream changes.[9][24] Medications for depression affect the transmission of serotonin, norepinephrine, and dopamine.[9] Older and less selective antidepressants like TCAs and MAOIs inhibit the reuptake or metabolism of norepinephrine and serotonin in the brain, which results in higher concentrations of neurotransmitters.[24] Antidepressants that have dual mechanisms of action inhibit the reuptake of both serotonin and norepinephrine and, in some cases, inhibit with weak effect the reuptake of dopamine.[9] Antidepressants affect variable neuronal receptors such as muscarinic cholinergic, α1- and α2-adrenergic, H1-histaminergic, and

sodium channels in the cardiac muscle, leading to decreased cardiac conduction and cardiotoxicity associated particularly with TCAs, and to a lesser extent with SSRIs.[25] Selectivity of antidepressant agents are based on the neurotransmitters that are thought to influence symptoms of depression.[26]
Drugs that selectively block the reuptake of serotonin and norepinephrine effectively treat depression and are better tolerated than TCAs. TCAs have comprehensive effects on various neurotransmitters receptors, which leads to lack of tolerability and increased risk of toxicity.[27]

Tricyclic antidepressants

Inhibiting the reuptake transport protein results in increased concentrations of serotonin and norepinephrine in the synaptic clefts, leading to improvement of depression symptoms.

TCAs were the first medications that had dual mechanism of action. The mechanism of action of tricyclic secondary amine antidepressants is only partly understood. TCAs have dual inhibition effects on norepinephrine reuptake transporters and serotonin reuptake transporters. Increased norepinephrine and serotonin concentrations are obtained by inhibiting both of these transporter proteins. TCAs have substantially more affinity for norepinephrine reuptake proteins than the SSRIs. This is because of a formation of secondary amine TCA metabolites.[28][29]

In addition, the TCAs interact with

adrenergic receptors
. This interaction seems to be critical for increased availability of norepinephrine in or near the synaptic clefts. Actions of imipramine-like tricyclic antidepressants have complex, secondary adaptions to their initial and sustained actions as inhibitors of norepinephrine transport and variable blockade of serotonin transport.

Norepinephrine interacts with postsynaptic α and β adrenergic receptor subtypes and presynaptic α2 autoreceptors. The α2 receptors include presynaptic

cyclic AMP-mediated phosphorylation-activation of the enzyme.[29] α2 receptors also cause decreased intracellular cyclic AMP expression which results in smooth muscle relaxation or decreased secretion.[30]

TCAs activate a negative feedback mechanism through their effects on presynaptic receptors. One probable explanation for the effects on decreased neurotransmitter release is that, as the receptors activate, inhibition of neurotransmitter release occurs (including suppression of voltage-gated Ca2+ currents and activation of G protein-coupled receptor-operated K+ currents). Repeated exposure of agents with this type of mechanism leads to inhibition of neurotransmitter release, but repeated administration of TCAs finally leads to decreased responses by α2 receptors. The desensitization of these responses may be due to increased exposure to endogenous norepinephrine or from the prolonged occupation of the norepinephrine transport mechanisms (via an allosteric effect). The adaptation allows the presynaptic synthesis and secretion of norepinephrine to return to, or even exceed, normal levels of norepinephrine in the synaptic clefts. Overall, inhibition of norepinephrine reuptake induced by TCAs leads to decreased rates of neuron firing (mediated through α2 autoreceptors), metabolic activity, and release of neurotransmitters.[29]

TCAs do not block dopamine transport directly but might facilitate dopaminergic effects indirectly by inhibiting dopamine transport into noradrenergic terminals of the cerebral cortex.[29] Because they affect so many different receptors, TCAs have adverse effects, poor tolerability, and an increased risk of toxicity.[27]

Selective serotonin reuptake inhibitors

Selective serotonin reuptake inhibitors (SSRIs) selectively inhibit the reuptake of serotonin and are a widely used group of antidepressants.

caudal raphe into the spinal cord
.

Selective norepinephrine reuptake inhibitors

Noradrenergic neurons are located in two major regions in the brain. These regions are

lateral tegmental. With administration of SNRIs, neuronal activity in locus coeruleus region is induced because of increased concentration of norepinephrine in the synaptic cleft. This results in activation of α2 adrenergic receptors,[24] as discussed previously
.

Assays have shown that SNRIs have insignificant penchant for mACh, α1 and α2 adrenergic, or H1 receptors.[26]

Dual serotonin and norepinephrine reuptake inhibitors

Agents with dual serotonin and norepinephrine reuptake inhibition (SNRIs) are sometimes called non-tricyclic serotonin and norepinephrine reuptake inhibitors. Clinical studies suggest that compounds that increase the concentration in the synaptic cleft of both norepinephrine and serotonin are more successful than single acting agents in the treatment of depression, but the data is not conclusive whether SNRIs are a more effective treatment option over SSRIs for depression.[32][33][34]

The non-tricyclic SNRIs have several important differences that are based on pharmacokinetics, metabolism to active metabolites, inhibition of CYP isoforms, effect of drug-drug interactions, and the half-life of the nontricyclic SNRIs.[28][35]

Combination of mechanisms of action in a single active agent is an important development in psychopharmacology.[35]

Structure activity relationship

Aryloxypropanamine scaffold

Several reuptake inhibitors contain an aryloxypropanamine scaffold. This structural motif has potential for high affinity binding to biogenic amine transports.

aromatic substituent also has a significant influence on the activity and selectivity of the compounds as inhibitors of the serotonin or the norepinephrine transporters.[35]

Aryloxypropanamine scaffold
Aryloxypropanamine scaffold and agents containing it.

Cycloalkanol ethylamine scaffold

norepinephrine and dopamine reuptake inhibition. Further synthesis and testing identified WAY-256805, a potent norepinephrine reuptake inhibitor that exhibited excellent selectivity and was efficacious in animal models of depression, pain, and thermoregulatory dysfunction.[37]

Cycloalkanol ethylamine scaffold
Cycloalkanol ethylamine scaffold and agents containing it.

Milnacipran

Structure of milnacipran.

Milnacipran is structurally different from other SNRIs.

allylic double bond showed significant improved effect on both norepinephrine and serotonin transporters.[38] Studies show that introducing a 2-methyl group in substituent R3, the potency at norepinephrine and serotonin transporters are almost abolished. Methyl groups in substituent groups R1 and R2 also abolish the potency at norepinephrine and serotonin transporters. Researchers found that replacing one of the ethyl groups of milnacipran with an allyl moiety increases the norepinephrine potency.[39] The pharmacophore of milnacipran derivatives is still largely unclear.[38]

The conformation of milnacipran is an important part of its pharmacophore. Changing its

molecular weight and low lipophilicity. Because of these properties, milnacipran exhibits almost ideal pharmacokinetics in humans such as high bioavailability, low inter-subject variability, limited liver enzyme interaction, moderate tissue distribution and a reasonably long elimination half-life. Milnacipran's lack of drug-drug interactions via cytochrome P450 enzymes is thought to be an attractive feature because many of the central nervous system drugs are highly lipophilic and are mainly eliminated by liver enzymes.[39]

Future development of structure activity relationship

The application of an aryloxypropanamine scaffold has generated a number of potent MAOIs.[40] Before the development of duloxetine, the exploration of aryloxypropanamine structure activity relationships resulted in the identification of fluoxetine and atomoxetine. The same motif can be found in reboxetine where it is constrained in a morpholine ring system. Some studies have been made where the oxygen in reboxetine is replaced by sulfur to give arylthiomethyl morpholine. Some of the arylthiomethyl morpholine derivatives maintain potent levels of serotonin and norepinephrine reuptake inhibition. Dual serotonin and norepinephrine reuptake inhibition resides in different enantiomers for arylthiomethyl morpholine scaffold.[41] Possible drug candidates with dual serotonin and norepinephrine reuptake inhibitory activity have also been derived from piperazine, 3-amino-pyrrolidine and benzylamine templates.[42]

Clinical trials

Depression

Several studies have shown that antidepressant drugs which have combined serotonergic and noradrenergic activity are generally more effective than SSRIs, which act upon serotonin reuptake by itself. Serotonergic-noradrenergic antidepressant drugs may have a modest efficacy advantage compared to SSRIs in treating major depressive disorder (MDD),[43] but are slightly less well tolerated.[44] Further research is needed to examine the possible differences of efficacy in specific MDD sub-populations or for specific MDD symptoms, between these classes of antidepressant drugs.

Analgesic

Data from clinical trials have indicated that SNRIs might have pain relieving properties. Although the perception and transmission of pain stimuli in the central nervous system have not been fully elucidated, extensive data support a role for serotonin and norepinephrine in the modulation of pain. Findings from clinical trials in humans have shown these antidepressants can help to reduce pain and functional impairment in central and neuropathic pain conditions. This property of SNRIs might be used to reduce doses of other pain relieving medication and lower the frequency of safety, limited efficacy and tolerability issues.[45] Clinical research data have shown in patients with GAD that the SNRI duloxetine is significantly more effective than placebo in reducing pain-related symptoms of GAD, after short-term and long-term treatment. However, findings suggested that such symptoms of physical pain reoccur in relapse situations, which indicates a need for ongoing treatment in patients with GAD and concurrent painful physical symptoms.[46]

Indications

SNRIs have been tested for treatment of the following conditions:

Pharmacology

Route of administration

SNRIs are delivered orally, usually in the form of capsules or tablets. It is recommended to take SNRIs in the morning with breakfast, which does not affect drug levels, but may help with certain side effects.[48] Norepinephrine has activating effects in the body and therefore can cause insomnia in some patients if taken at bedtime.[49] SNRIs can also cause nausea, which is usually mild and goes away within a few weeks of treatment, but taking the medication with food can help alleviate this.[50]

Mode of action

The condition for which SNRIs are mostly indicated, major depressive disorder, was thought to be mainly caused by decreased levels of serotonin and

tricyclic antidepressants (TCAs), but not with the SSRIs.[53]

Recent studies have shown that depression may be linked to increased inflammatory response,[54] thus attempts at finding an additional mechanism for SNRIs have been made. Studies have shown that SNRIs as well as SSRIs have significant anti-inflammatory action on microglia[55] in addition to their effect on serotonin and norepinephrine levels. As such, it is possible that an additional mechanism of these drugs that acts in combination with the previously understood mechanism exist. The implication behind these findings suggests use of SNRIs as potential anti-inflammatories following brain injury or any other disease where swelling of the brain is an issue. However, regardless of the mechanism, the efficacy of these drugs in treating the diseases for which they have been indicated has been proven, both clinically and in practice.[improper synthesis?]

Pharmacodynamics

Most SNRIs function alongside

frontal cortex, an area of the brain largely lacking in dopamine transporters. This effect of SNRIs results in increased dopamine neurotransmission, in addition to the increases in serotonin and norepinephrine activity.[56] Furthermore, because SNRIs are extremely selective, they have no measurable effects on other, unintended receptors, in contrast to monoamine oxidase inhibition.[57] Pharmaceutical tests have determined that use of both SNRIs or SSRIs can generate significant anti-inflammatory action on microglia, as well.[55][15][58][59][10][60]

Activity profiles

SNRIs at the human SERT and NET[61][62][6][63]
Compound SERT NET ~Ratio
(5-HT : NE)
Ki IC50 Ki IC50
Venlafaxine 7.8 145 1,920 1420 9.8:1
Des-venlafaxine
40.2 47.3 558.4 531.3
Duloxetine 0.07 3.7 1.17 20 5.4:1
Atomoxetine 87[62] 5.4[62] 0.06:1
(= 1:16)
Milnacipran 8.44 151 22 68 0.45:1 (= 1:2.2)
Levo-milnacipran
11.2 19.0 92.2 10.5
All of the Ki and IC50 values are nM. The 5-HT/NE ratio is
based on IC50 values for the SERT and NET.[61]

Pharmacokinetics

The half-life of venlafaxine is about 5 hours, and with once-daily dosing, steady-state concentration is achieved after about 3 days, though its active metabolite desvenlafaxine lasts longer.[59] The half-life of desvenlafaxine is about 11 hours, and steady-state concentrations are achieved after 4 to 5 days.[58] The half-life of duloxetine is about 12 hours (range: 8–17 hours), and steady-state is achieved after about 3 days.[10] Milnacipran has a half-life of about 6 to 8 hours, and steady-state levels are reached within 36 to 48 hours.[60]

Contraindications

SNRIs are contraindicated in patients taking

LSD.[64] Signs and symptoms of serotonin syndrome include: hyperthermia, rigidity, myoclonus, autonomic instability with fluctuating vital signs, and mental status changes that include extreme agitation progressing to delirium and coma.[10]

Due to the effects of increased

noradrenergic activity, pre-existing hypertension should be controlled before treatment with SNRIs and blood pressure periodically monitored throughout treatment.[65] Duloxetine has also been associated with cases of liver failure and should not be prescribed to patients with chronic alcohol use or liver disease. Studies have found that Duloxetine can increase liver function tests three times above their upper normal limit.[66] Patients with coronary artery disease should caution the use of SNRIs.[67] Furthermore, due to some SNRIs' actions on obesity, patients with major eating disorders such as anorexia nervosa or bulimia should not be prescribed SNRIs.[15] Duloxetine and milnacipran are also contraindicated in patients with uncontrolled narrow-angle glaucoma, as they have been shown to increase incidence of mydriasis.[10][60]

Side effects

Because the SNRIs and SSRIs act in similar ways to elevate serotonin levels, they share many side effects, though to varying degrees. The most common side effects include nausea/vomiting, sweating, loss of appetite, dizziness, headache, increase in suicidal thoughts, and sexual dysfunction.[68] Elevation of norepinephrine levels can sometimes cause anxiety, mildly elevated pulse, and elevated blood pressure. However, norepinephrine-selective antidepressants, such as reboxetine and desipramine, have successfully treated anxiety disorders.[69] People at risk for hypertension and heart disease should monitor their blood pressure.[15][58][59][10][60]

Sexual Dysfunction

SNRIs, similarly to SSRIs, can cause several types of sexual dysfunction, such as erectile dysfunction, decreased libido, sexual anhedonia, and anorgasmia.[10][59][70] The two common sexual side effects are diminished interest in sex (libido) and difficulty reaching climax (anorgasmia), which are usually somewhat milder with SNRIs compared to SSRIs.[71] To manage sexual dysfunction, studies have shown that switching to or augmenting with bupropion or adding a PDE5 Inhibitor have decreased symptoms of sexual dysfunction.[72] Studies have shown that PDE5 Inhibitors, such as sildenafil (Viagra), tadalafil (Cialis), vardenafil (Levitra), and avanafil (Stendra), have sometimes been helpful to decrease the sexual dysfunction, including erectile dysfunction, although they have been shown to be more effective in men than women.[72]

Serotonin Syndrome

A serious, but rare, side effect of SNRIs is serotonin syndrome, which is caused by an excess of serotonin in the body. Serotonin syndrome can be caused by taking multiple serotonergic drugs, such as SSRIs or SNRIs. Other drugs that contribute to serotonin syndrome include MAO inhibitors, linezolid, tedizolid, methylene blue, procarbazine, amphetamines, clomipramine, and more.[73] Early symptoms of serotonin syndrome may include nausea, vomiting, diarrhea, sweating, agitation, confusion, muscle rigidity, dilated pupils, hyperthermia, rigidity, and goose bumps. More severe symptoms include fever, seizures, irregular heartbeat, delirium, and coma.[74][75][10] If signs or symptoms arise, discontinue treatment with serotonergic agents immediately.[74] It is recommended to washout 4 to 5 half-lives of the serotonergic agent before using an MAO inhibitor.[76]

Bleeding

Some studies suggest there are risks of upper gastrointestinal bleeding, especially venlafaxine, due to impairment of platelet aggregation and depletion of platelet serotonin levels.[77][78] Similarly to SSRIs, SNRIs may interact with anticoagulants, like warfarin. There is more evidence of SSRIs having higher risk of bleeding than SNRIs.[77] Studies have suggested caution when using SNRIs or SSRIs with high doses of nonsteroidal anti-inflammatory drugs (NSAIDs), such as ibuprofen or naproxen due to an increased risk of upper GI bleeding.[34]

Visual problems

Image depicting the vision of someone with visual snow syndrome or hallucinogen persisting perception disorder.

Similarly to other antidepressants, SNRI medications have been found to cause visual snow syndrome, a condition characterized by visual static, palinopsia (negative after image), nyctalopia (poor vision at night), and photophobia (brighter presentation of lights or highlighted colors). Evidence shows that 8.9% of those taking SNRIs experienced visual snow, 10.5% experienced palinopsia, 15.3% experienced photophobia, and 17.7% experienced nyctolopia as the result of SNRI prescription intake. Amitriptyline, and citalopram have also been reported to worsen or cause symptoms of VSS.[79]

Precautions

Starting an SNRI regimen

Due to the extreme changes in noradrenergic activity produced from norepinephrine and serotonin reuptake inhibition, patients that are just starting an SNRI regimen are usually given lower doses than their expected final dosing to allow the body to acclimate to the drug's effects. As the patient continues along at low doses without any side-effects, the dose is incrementally increased until the patient sees improvement in symptoms without detrimental side-effects.[80]

Discontinuation syndrome

As with SSRIs, the abrupt discontinuation of an SNRI usually leads to

discontinuation syndrome", which could include states of anxiety and other symptoms. Therefore, it is recommended that users seeking to discontinue an SNRI slowly taper the dose under the supervision of a professional. Discontinuation syndrome has been reported to be markedly worse for venlafaxine when compared to other SNRIs. As such, as tramadol is related to venlafaxine, the same conditions apply.[81] This is likely due to venlafaxine's relatively short half-life and therefore rapid clearance upon discontinuation. In some cases, switching from venlafaxine to fluoxetine, a long-acting SSRI, and then tapering off fluoxetine, may be recommended to reduce discontinuation symptoms.[82][83] Signs and symptoms of withdrawal from abrupt cessation of an SNRI include dizziness, anxiety, insomnia, nausea, sweating, and flu-like symptoms, such as lethargy and malaise.[84]

Overdose

Causes

Overdosing on SNRIs can be caused by either drug combinations or excessive amounts of the drug itself. Venlafaxine is marginally more toxic in overdose than duloxetine or the SSRIs.[15][58][59][10][60][85]

Symptoms

Symptoms of SNRI overdose, whether it be a mixed drug interaction or the drug alone, vary in intensity and incidence based on the amount of medicine taken and the individuals sensitivity to SNRI treatment. Possible symptoms may include:[10]

Management

Overdose is usually treated symptomatically, especially in the case of serotonin syndrome, which requires treatment with cyproheptadine and temperature control based on the progression of the serotonin toxicity.[86] Patients are often monitored for vitals and airways cleared to ensure that they are receiving adequate levels of oxygen. Another option is to use activated carbon in the GI tract in order to absorb excess neurotransmitter.[10]

Comparison to SSRIs

Because SNRIs were developed more recently than SSRIs, there are relatively few of them. However, the SNRIs are among the most widely used antidepressants today. In 2009, Cymbalta and Effexor were the 11th- and 12th-most-prescribed branded drugs in the United States, respectively. This translates to the 2nd- and 3rd-most-common antidepressants, behind Lexapro (escitalopram), an SSRI.[87] In some studies, SNRIs demonstrated slightly higher antidepressant efficacy than the SSRIs (response rates 63.6% versus 59.3%).[43] However, in one study escitalopram had a superior efficacy profile to venlafaxine.[88]

Special populations

Pregnancy

No antidepressants are FDA approved during pregnancy.[89] Use of antidepressants during pregnancy may result in fetus abnormalities affecting functional development of the brain and behavior.[89]

Pediatrics

SSRIs and SNRIs have been shown to be effective in treating major depressive disorder and anxiety in pediatric populations.[90] However, there is a risk of increased suicidality in pediatric populations for treatment of major depressive disorder, especially with venlafaxine.[90] Fluoxetine is the only antidepressant that is approved for child/adolescent major depressive disorder.[91]

Geriatrics

Most antidepressants, including SNRIs, are safe and effective in the geriatric population. Decisions are often based on co-morbid conditions, drug interactions, and patient tolerance. Due to differences in body composition and metabolism, starting doses are often half that of the recommended dose for younger adults.[92]

Research

A systematic review that looked at the efficacy of antidepressants for pain relief concludes that only 11 of 42 comparisons showed evidence of efficacy. Seven of the eleven comparisons belong to the SNRI drug class.[93]

See also

References

  1. ^ Cleveland Clinic medical professional (2023-03-05). "SNRIs". Cleveland Clinic. Retrieved 2024-01-09.
  2. PMID 19740668
    .
  3. .
  4. .
  5. .
  6. ^ .
  7. ^ .
  8. .
  9. ^ .
  10. ^ a b c d e f g h i j k l "Cymbalta- duloxetine hydrochloride capsule, delayed release". DailyMed. 20 September 2021. Retrieved 12 February 2023.
  11. ^ "Yentreve (duloxetine hydrochloride) Hard Gastro-Resistant Capsules. Summary of Product Characteristics" (PDF). European Medicines Agency. Archived from the original (PDF) on 26 June 2016. Retrieved 29 August 2016.
  12. ^ "Nefazodone hydrochloride tablet". DailyMed. 16 November 2021. Retrieved 12 February 2023.
  13. ^
    S2CID 22242897
    .
  14. ^ .
  15. ^ a b c d e "Meridia (sibutramine hydrochloride monohydrate) Capsules C-IV. Full Prescribing Information (archived label)". Abbott Laboratories, North Chicago, IL 60064, USA. Retrieved 2 September 2016.
  16. PMID 12007530
    .
  17. ^ Rockoff JD, Dooren JC (October 8, 2010). "Abbott Pulls Diet Drug Meridia Off US Shelves". The Wall Street Journal. Archived from the original on 11 October 2010. Retrieved 8 October 2010.
  18. S2CID 22620947
    .
  19. ^ .
  20. ^ .
  21. .
  22. ^ a b Lieberman JA (2003). "History of the Use of Antidepressants in Primary Care" (PDF). Primary Care Companion J Clin Psychiatry. 5 (S7): 6–10. Archived from the original (PDF) on 2014-06-11. Retrieved 2014-03-05.
  23. .
  24. ^ .
  25. .
  26. ^ .
  27. ^ .
  28. ^ a b c Lemke TL, Williams DA, Roche VF, Zito SW (2008). Foye's principles of medicinal chemistry (6th ed.). USA: Lippincott Williams & Wilkins. pp. 547–67, 581–582.
  29. ^ a b c d e Brunton, L.L., Lazo, J.S., Parker, K.L., eds. (2006). Goodman & Gilman's: The Pharmacological Basis of Therapeutics (11 ed.). New York: McGraw-Hill.
  30. ^ Silverthorn, D.U., ed. (2007). Human Physiology (4 ed.). San Francisco: Pearson. pp. 383–384.
  31. S2CID 23634771
    .
  32. .
  33. .
  34. ^ .
  35. ^ .
  36. ^ .
  37. .
  38. ^ .
  39. ^ .
  40. .
  41. .
  42. .
  43. ^ .
  44. .
  45. .
  46. .
  47. .
  48. .
  49. .
  50. ^ "Helpful for chronic pain in addition to depression". Mayo Clinic. Retrieved 2019-10-24.
  51. S2CID 250646781
    .
  52. .
  53. .
  54. .
  55. ^ .
  56. ^ "Cambridge University Press - Service Announcement".
  57. S2CID 37792842
    .
  58. ^ a b c d "Pristiq Extended-Release- desvenlafaxine succinate tablet, extended release". DailyMed. 25 March 2022. Retrieved 12 February 2023.
  59. ^ a b c d e "Effexor XR- venlafaxine hydrochloride capsule, extended release". DailyMed. 29 August 2022. Retrieved 12 February 2023.
  60. ^ a b c d e "Savella- milnacipran hydrochloride tablet, film coated Savella- milnacipran hydrochloride kit". DailyMed. 23 December 2022. Retrieved 12 February 2023.
  61. ^
    PMID 28503727
    .
  62. ^ .
  63. ^ Roth BL, Driscol J (Dec 2012). "PDSP Ki Database". Psychoactive Drug Screening Program (PDSP). University of North Carolina at Chapel Hill and the United States National Institute of Mental Health. Retrieved 7 July 2018.
  64. ^
    S2CID 37959124
    .
  65. .
  66. ^ McIntyre RS et al. The hepatic safety profile of duloxetine: a review. Expert Opin Drug Metab Toxicol. 2008;4(3):281–285.
  67. PMID 29315692
    .
  68. ^ "SNRI Antidepressants". poison.org. Retrieved 2019-10-21.
  69. PMID 11838623
    .
  70. .
  71. .
  72. ^ .
  73. ^ "Serotonin syndrome: Preventing, recognizing, and treating it". www.mdedge.com. Retrieved 2019-11-21.
  74. ^
    PMID 18625822
    .
  75. ^ "SNRI Antidepressants". poison.org. Retrieved 2019-10-23.
  76. S2CID 145217439
    .
  77. ^ .
  78. .
  79. .
  80. ^ "Duloxetine: Drug Information". UpToDate. Retrieved 28 June 2012.
  81. PMID 17445831
    .
  82. .
  83. .
  84. .
  85. .
  86. . Retrieved 2019-11-21.
  87. ^ "2009 Top 200 branded drugs by total prescriptions" (PDF). SDI/Verispan, VONA, full year 2009. www.drugtopics.com. Archived from the original (PDF) on 14 July 2011. Retrieved 6 April 2011.
  88. PMID 17394446
    .
  89. ^ .
  90. ^ .
  91. .
  92. .
  93. .