Neuromodulation: An Overview

Neuromodulation is a treatment strategy that modifies neuronal activity by precisely delivering chemical or electrical stimulation to particular nervous system regions. This method can potentially treat several neurological and psychiatric conditions by restoring or modifying nerve function.

Historical Background and Development

Ancient civilizations are where neuromodulation first emerged. Although the details of the Nile catfish's medical use are still unknown, the Egyptians were among the first to notice its electrical qualities. Philosophers such as Plato and Aristotle recorded the usage of electric fish discharges to cure illnesses in ancient Greece. An early example of electrical neuromodulation was torpedo fish applied to the scalp to treat migraines, as the Roman physician Scribonius Largus recounted.

Significant progress was made in the 18th century with the development of the Leyden jar and electrostatic machines, which allowed for the regulated application and storage of electricity. Thanks to the creation of the Galvanic battery by Italian scientist Luigi Galvani, direct current (DC) stimulation was first used therapeutically for diseases, including major depression. However, as electroconvulsive therapy (ECT) gained popularity in the 1930s, DC stimulation research began to wane due to inconsistent results and a lack of understanding of the mechanisms involved.

Deep brain stimulation (DBS) for chronic, intractable pain marked the beginning of the current era of neuromodulation in the early 1960s. An important turning point in the treatment of pain was reached in 1967 when Dr. Norman Shealy inserted the first spinal cord stimulator. Melzack and Wall's 1965 Gate Control Theory of Pain, which postulated that non-painful input might inhibit pain perceptions, impacted these advancements. Since then, neuromodulation has broadened to encompass a variety of methods, including transcranial magnetic stimulation (TMS), spinal cord stimulation (SCS), and vagus nerve stimulation (VNS). Chronic pain, epilepsy, depression, and Parkinson's disease have all been treated with these techniques. Thanks to technological advancements, patients now have more accessible treatment options thanks to the creation of non-invasive gadgets like brain-stimulation headsets.

Closed-loop systems that offer real-time feedback have emerged in the field in recent years, improving treatment accuracy and effectiveness. There is continuous research into new uses, such as neuromodulation for autoimmune diseases and inflammatory bowel disease. Neuromodulation keeps developing as our knowledge of neural networks expands, providing exciting new treatment options for a variety of ailments.

Mechanism of Neuromodulation

Neuromodulation is the process of controlling neuronal activity using neurotransmitters and neuromodulators, which are essential for adjusting synaptic plasticity and neuronal excitability.

Role of Neurotransmitters and Neuromodulators

Neurotransmitters that modulate synaptic transmission and plasticity include acetylcholine, serotonin, and dopamine.

Dopamine: This neurotransmitter is essential for reward systems, behavior reinforcement, emotional control, and motor control. Dopamine has a well-established impact on synaptic plasticity, especially on the cellular processes that underlie learning and memory, long-term potentiation (LTP), and long-term depression (LTD).

Serotonin: Through its effects on presynaptic and postsynaptic processes, serotonin regulates synaptic plasticity. Neuronal excitability and synaptic strength may be impacted by changes in neurotransmitter release and receptor sensitivity. These modulatory effects are essential for regulating emotions and many cognitive processes.

Acetylcholine: Acetylcholine plays a significant role in attention, learning, and memory. It modulates synaptic plasticity by affecting the induction and maintenance of LTP and LTD. Acetylcholine's action on muscarinic and nicotinic receptors influences neuronal excitability and synaptic transmission, thereby contributing to cognitive processes.

Impact on Neuronal Excitability and Synaptic Plasticity

Neuromodulators influence synaptic plasticity and neuronal excitability in several ways:

Presynaptic Modulation: Neuromodulators can change the likelihood of neurotransmitter release from presynaptic terminals. For example, they can alter synaptic strength by activating presynaptic receptors, which can either increase or decrease neurotransmitter release.

Postsynaptic Modulation: Neuromodulators can affect the response of postsynaptic neurons by altering ion channel activity or receptor sensitivity. This modulation may impact neuronal excitability and synaptic plasticity, modifying the magnitude and duration of postsynaptic potentials.

Glial Cell Interaction: Glial cells, which are involved in preserving synaptic homeostasis and regulating synaptic transmission, interact with neuromodulators. This relationship may impact synaptic plasticity and the general operation of brain networks.

Types of Neuromodulation

Neuromodulation refers to a variety of methods for treating neurological and mental disorders by changing nerve activity. These techniques fall into three general categories: noninvasive, chemical, and electrical.

1. Electrical Neuromodulation

For modifying brain activity, electrical stimuli are applied. Important methods consist of:

Deep Brain Stimulation (DBS): To provide regulated electrical pulses, DBS involves implanting electrodes into particular brain areas. DBS is thought to affect both local and network-wide brain activity, modifying oscillatory patterns and encouraging synaptic plasticity, while the precise mechanisms are still being studied. Depending on the illness being treated and the area of the brain being addressed, these effects can differ.

Spinal Cord Stimulation (SCS): To treat chronic pain, SCS entails inserting electrodes into the spinal cord's epidural area. Pain alleviation may result from the stimulation's primary activation of the dorsal cord's large-diameter sensory afferents.

Peripheral Nerve Stimulation (PNS): PNS uses electrical impulses to target particular peripheral nerves to control pain or restore function. In clinical practice, this approach is applied to ailments such as neuropathic pain.

2. Chemical Neuromodulation

This method modifies brain activity by either pharmacological agents or genetic interventions:

Pharmacological Approaches: Neuromodulatory drugs can affect neuronal circuits by changing receptor activation or neurotransmitter levels. For instance, drugs that target dopamine pathways are used to treat Parkinson's disease.

Gene Therapy: This method entails putting genetic material into cells to make up for damaged genes or create useful proteins. Treatment of genetic problems at the level of malfunctioning cells, such as retinal cells in certain eye diseases, is the goal of gene therapy in the context of neuromodulation.

Optogenetics: Optogenetics uses light to regulate the activity of certain neurons in living tissue by combining genetic and optical techniques. To do this, genes expressing light-sensitive ion channels are inserted into target neurons, enabling precise temporal regulation of their activity.

3. Non-Invasive Neuromodulation

These methods alter brain function without requiring surgery:

Transcranial Magnetic Stimulation (TMS): TMS stimulates brain nerve cells using magnetic fields. The U.S. Food and Drug Administration (FDA) has approved this non-invasive therapy to treat serious depression, particularly in cases where other treatments have failed.

Transcranial Direct Current Stimulation (tDCS): Through electrodes, tDCS modifies neuronal activity by delivering a small electrical current to the scalp. tDCS has the potential to be a therapeutic benefit for several illnesses since it can either excite or inhibit neuronal activity, depending on the polarity of the current.

Pharmacological Neuromodulators

Pharmacological neuromodulators are compounds that change the release of neurotransmitters, the sensitivity of receptors, or the transmission of nerve signals. They are essential in the treatment of mental and neurological disorders such as epilepsy, depression, Parkinson's disease, and chronic pain. In addition to injectable neuromodulators like botulinum toxin (Botox, Dysport, Xeomin, Jeuveau), which block nerve signals to lessen muscle spasticity, migraines, dystonia, and excessive perspiration, these agents also include medications that control neurotransmitter systems, such as dopamine, serotonin, GABA, and glutamate. Neuromodulators offer efficient therapy solutions that enhance patient outcomes and quality of life by specifically addressing brain pathways. Following are different neuromodulators that are being used for treating several conditions:

1. Botulinum Toxins (Local Neuromodulators)

Strong neuromodulators like botulinum toxins offer both medicinal and cosmetic advantages by momentarily blocking nerve signals to muscles and glands. These toxins, which are derived from Clostridium botulinum, function by preventing the release of acetylcholine, a neurotransmitter that causes muscles to contract. Because of this process, botulinum toxins are very useful in the treatment of diseases like hyperhidrosis (excessive perspiration), muscle stiffness, persistent headaches, and dystonia. Formulations like Botox, Dysport, Xeomin, and Jeuveau are used extensively in cosmetic dermatology to minimize wrinkles and fine lines in addition to their medicinal uses. Botulinum toxins have transformed the treatment of neuromuscular and autonomic disorders by providing focused, reversible neuromodulation.

  1. Botox (Botulinum toxin type A)

The bacterium Clostridium botulinum produces the neurotoxic protein known as botulinum toxin type A or Botox. By preventing the release of neurotransmitters, it is frequently utilized in the medical and cosmetic domains to momentarily lower muscle activity.

Fact

Botox has been allowed to treat several medical diseases in addition to its cosmetic application of reducing wrinkles on the face. These conditions include cervical dystonia, overactive bladder, chronic migraines, and excessive perspiration (hyperhidrosis).

Mechanism of Action

Acetylcholine (ACh), a neurotransmitter necessary for muscular contraction and autonomic nervous system processes, is blocked by botulinum toxin type A (Botox). This is accomplished using the multi-step procedure listed below:

Binding – At the neuromuscular junction, Botox binds specifically to high-affinity receptors on presynaptic cholinergic nerve terminals.

Internalization – The toxin forms a vesicle inside the nerve cell after being absorbed into the neuron through receptor-mediated endocytosis.

Translocation – After entering, Botox's light chain is discharged into the cytoplasm, where it functions as an enzyme that breaks down proteins.

SNAP-25 Cleavage – A vital part of the SNARE protein complex, SNAP-25 is cleaved by Botox. Acetylcholine cannot be released into the synaptic cleft as a result of the synaptic vesicles' inability to fuse with the nerve terminal membrane.

Muscle Paralysis – Temporary muscular paralysis or relaxation can occur when acetylcholine is depleted because the targeted muscle cannot receive signals to contract.

Duration of Effect

After three to four months, the benefits of Botox injections usually wear off, and muscular function gradually resumes, requiring more treatments to keep the results.

Clinical Data on Effectiveness

Botulinum toxin type A (BoNT-A) has been shown in clinical trials to be effective in treating several illnesses. Botox dramatically lowers the number of headache days and hours experienced by chronic migraine sufferers each month. It works well for managing spasticity, which includes reducing stiffness and increasing muscular tone, especially in people with neurological conditions. Furthermore, with long-lasting analgesic benefits over 24 weeks, BoNT-A has demonstrated promise in the treatment of peripheral neuropathic pain. Additionally, studies indicate that it may help patients with severe vasospastic symptoms by enhancing blood flow in Raynaud's phenomenon, which is a side effect of scleroderma.

Other aesthetic uses of BoNT-A have been investigated. Injecting the toxin into overactive upper lip muscles, for example, has been used to treat a "gummy smile" by lowering excessive gingival show. BoNT-A has also been used to contour the lower face, especially in minimizing hypertrophy of the masseter muscles, which can narrow the jawline. For lower face rejuvenation, recent clinical trials have also examined the use of BoNT-A in conjunction with hyaluronic acid fillers, with superior outcomes than either treatment alone. These studies demonstrate the adaptability of BoNT-A in aesthetic medicine by emphasizing how it can be used to accomplish desired cosmetic results through both muscle and neuromodulation.

Dosage Forms Available

Botox is provided as a sterile, vacuum-dried powder for reconstitution. Each vial contains a predetermined quantity of units of botulinum toxin type A, human albumin, and sodium chloride, which serve as stabilizers. After being reconstituted with sterile saline, the solution is prepared for intramuscular or intradermal injection.

Other Clinical Benefits

In addition to its main indications, Botox has been investigated for several off-label applications, including the treatment of temporomandibular joint abnormalities, excessive drooling, and some forms of neuropathic pain. Research on its potential in various medical disorders is also ongoing. It includes the use of Botox in spasticity management, postoperative atrial fibrillation prevention, and Raynaud's phenomenon secondary to scleroderma.

Side Effects Associated

Localized discomfort, bruising, or swelling at the injection site are typical Botox injection adverse effects. Some patients might have flu-like symptoms or headaches. Rarely, the effects of the toxin may extend past the injection site and cause symptoms including swallowing difficulties, eye issues, or muscle weakness.

Administration Dose and Guidelines

The condition being treated, the particular muscles affected, and the patient's unique circumstances all influence the right dosage of Botox.

Contraindications

People who have a history of known hypersensitivity to any preparation of botulinum toxin or any of the ingredients in the formulation should not use Botox. Patients who have active infections at the suggested injection sites shouldn't receive it. Patients with neuromuscular diseases should also exercise caution since they may be more susceptible to systemic effects.

  1. Jeuveau (PrabotulinumtoxinA-xvfs)

Jeuveau, a type A botulinum toxin product created specifically for cosmetic purposes, is scientifically known as prabotulinumtoxinA-xvfs. The U.S. Food and Drug Administration (FDA) approved Jeuveau, a product made by Evolus, in February 2019 for the short-term treatment of moderate to severe glabellar lines, also referred to as frown lines, in adults.

Fact

Unlike other botulinum toxin products with both medical and aesthetic indications, Jeuveau is the first to be produced only for cosmetic purposes.

Mechanism of Action

Jeuveau works by preventing acetylcholine from being released at the neuromuscular junction. By blocking nerve signals from reaching the targeted muscles, this inhibition temporarily relaxes the muscles and lessens the visibility of dynamic wrinkles.

Clinical Data on Effectiveness

When it came to treating moderate to severe glabellar lines (frown lines), the Phase III clinical trial assessing Jeuveau (prabotulinumtoxinA-xvfs) showed that it was not inferior to Botox. On Day 2, 54% of patients treated with Jeuveau showed a 1-grade improvement in their frown lines, which was substantially greater than the placebo group in this multicenter, randomized, double-blind, placebo-controlled research. Seventy percent of patients showed improvement by Day 30, demonstrating that the results were sustained. Common side effects like moderate headaches and eyelid ptosis that went away fast were part of the safety profile, which was similar to that of other botulinum toxin products. These results supported Jeuveau's FDA approval for cosmetic use by confirming its safety, effectiveness, and results that were equivalent to those of Botox.

Dosage Forms Available

Each single-use vial of Jeuveau, a sterile, lyophilized powder, contains 100 units of prabotulinumtoxinA-xvfs. Before delivery, 2.5 mL of an injection of 0.9% sodium chloride is used to reconstitute the powder.

Other Clinical Benefits

Jeuveau has been investigated for alternative cosmetic uses, such as the treatment of crow's feet and forehead lines, in addition to its core use for glabellar lines. These applications should be regarded as off-label, nevertheless, as the FDA has not approved them.

Side Effects Associated

Common adverse effects of Jeuveau include drooping eyelids, headaches, and cold-like feelings. Although they are less frequent, serious side effects can include neck pain, exhaustion, and dry mouth.

Administration Dose and Guidelines

Jeuveau is injected intramuscularly into each of the five locations in a volume of 0.1 mL (4 units) to treat glabellar lines. This results in a total of 20 units. The frequency of retreatment should not exceed every three months.

Contraindications

Patients who are known to be hypersensitive to prabotulinumtoxinA-xvfs or any of its constituents should not use Jeuveau. Additionally, it should not be administered to those who have an infection at the intended injection sites or who have neuromuscular conditions like Lambert-Eaton syndrome or myasthenia gravis.

2. Dopaminergic Neuromodulators

The pharmaceutical class known as dopaminergic modulators affects the action of dopamine, a crucial neurotransmitter that controls motivation, emotion, movement, and thought. These modulators are frequently used to treat neurological and psychiatric disorders such as Parkinson's disease, schizophrenia, and attention-deficit hyperactivity disorder (ADHD). They either increase or decrease dopamine transmission in the brain. To modify dopamine's availability and activity in the brain, dopaminergic modulators either act on dopamine receptors or change the synthesis, release, and reuptake of dopamine. These medications can help restore equilibrium to disturbed dopamine systems and reduce symptoms of illnesses connected to dopamine by focusing on particular dopaminergic pathways.

  1. Levodopa

Parkinson's disease and other types of parkinsonism are the main conditions that are treated with levodopa, commonly referred to as L-DOPA. It functions as a precursor to dopamine, a neurotransmitter that Parkinson's disease patients' brains lack in sufficient amounts. Levodopa reduces motor symptoms such as bradykinesia (slowness of movement), rigidity, and tremors by raising dopamine levels.

Fact

Carbidopa, a peripheral dopa decarboxylase inhibitor, is frequently used in conjunction with levodopa. Only a small amount of levodopa reaches the central nervous system when taken orally because it is quickly decarboxylated to dopamine in peripheral tissues. To increase its bioavailability, combo therapy must be used. For this reason, peripheral dopa decarboxylase inhibitors like carbidopa are frequently used in conjunction with levodopa.

Mechanism of Action

The brain's dopa decarboxylase enzyme transforms levodopa into dopamine when it passes through the blood-brain barrier. By restoring dopamine reserves, this conversion enhances dopaminergic neurotransmission and reduces Parkinson's disease-related motor symptoms.

Clinical Data on Effectiveness

Clinical data continuously support Levodopa's efficacy in reducing motor symptoms in Parkinson's disease. Research indicates that levodopa considerably enhances everyday activities, quality of life, and motor function in both the early and advanced phases of the illness. Early levodopa introduction yields significant motor gains without compromising long-term results, according to the LEAP research. Nevertheless, prolonged use may result in motor issues such as dyskinesias and the "on-off" phenomenon, necessitating therapy modifications. These results demonstrate the critical role levodopa plays in the treatment of Parkinson's disease, with careful monitoring required to balance side effects and efficacy.

Dosage Forms Available

There are several formulations of levodopa available, including:

Oral Tablets: Frequently used in conjunction with carbidopa to increase effectiveness and lessen ancillary side effects.

Oral Inhalation Powder: For Parkinson's patients already receiving levodopa and carbidopa therapy, oral inhalation powder is recommended for the sporadic treatment of off episodes.

Other Clinical Benefits

Levodopa is used to treat post-encephalitic parkinsonism and symptomatic parkinsonism after carbon monoxide overdose in addition to Parkinson's disease.

Side Effects Associated

Levodopa frequently causes the following side effects: orthostatic hypotension, hallucinations, confusion, dyskinesias (involuntary movements that might happen with prolonged use), nausea, and vomiting, which are frequently brought on by peripheral dopamine production.

Administration Dose and Guidelines

The needs of each patient and the intensity of their symptoms determine the levodopa dosage. To reduce side effects, it is usually started at a low dose and raised gradually.

Contraindications

Levodopa should not be used in patients who have:

  • Hypersensitivity: To any ingredient in the formulation, including levodopa.
  • The cause of narrow-angle glaucoma.
  • Melanoma: Levodopa may make melanoma more likely.
  • Are taking nonselective monoamine oxidase inhibitors (MAOIs) as the possibility of a hypertensive crisis, concurrent use
  • Levodopa should also be used with caution in patients who have a history of peptic ulcer disease, mental health issues, or cardiovascular illness.
  1. Pramipexole (Mirapex)

Pramipexole is a non-ergot dopamine agonist that is sold under the brand name Mirapex. It is used to treat the symptoms of RLS and idiopathic Parkinson's disease.

Fact

Pramipexole, which treats a variety of dopaminergic deficits, is licensed for both Parkinson's disease and restless legs syndrome.

Mechanism of Action

As a dopamine receptor agonist, pramipexole mainly targets the D₂ and D₃ subtypes, which are important for controlling mood and motor function. Pramipexole makes up for the decreased dopamine levels in the brain by attaching to and activating these dopamine receptors, especially in regions like the basal ganglia that are impacted by Parkinson's disease. This action reduces the discomfort and sleep difficulties associated with Restless Legs Syndrome as well as the motor signs of Parkinson's disease, including tremors, bradykinesia, and stiffness. Furthermore, it is believed that pramipexole's ability to selectively activate the D₃ receptor adds to its efficacy in treating these illnesses.

Clinical Data on Effectiveness

Based on clinical studies, pramipexole successfully reduces Parkinson's disease motor symptoms. Pramipexole dramatically enhanced motor function and quality of life in patients with early-stage Parkinson's disease, according to research published in Neurology in 2000.

Pramipexole has been demonstrated to reduce symptoms and enhance sleep quality in the treatment of restless legs syndrome. Pramipexole improved sleep efficiency and lessened the intensity of RLS symptoms based on another clinical study.

Dosage Forms Available

Both immediate-release and extended-release oral tablet forms of pramipexole are accessible. Extended-release tablets are usually taken once daily, but immediate-release tablets are usually taken three times a day.

Other Clinical Benefits

Pramipexole has been studied for its possible neuroprotective benefits in Parkinson's disease in addition to its main uses. Although further research is required to corroborate these findings, some studies indicate that it may halt the advancement of the disease.

Side Effects Associated

Orthostatic hypotension, nausea, dizziness, and somnolence are typical pramipexole side effects. Additionally, it could lead to obsessive behaviors like excessive shopping, hypersexuality, or gambling.

Administration Dose and Guidelines

The ailment being treated determines the pramipexole dosage:

Parkinson's Disease: The starting dosage is usually three doses of 0.375 mg daily. Depending on the patient's reaction and tolerance, the dosage may be progressively raised.

Restless Legs Syndrome: The suggested dosage for restless legs syndrome is 0.125 mg once a day, two to three hours before bedtime.

Patients with renal impairment require dose modifications.

Contraindications

Patients who are hypersensitive to pramipexole or any of its ingredients should not take it. Because it can make psychotic disorders worse, it should be used carefully in individuals who have a history of them.

3. Serotonergic Neuromodulators

  1. Fluoxetine (Prozac)

Prozac, also known as fluoxetine, is a selective serotonin reuptake inhibitor (SSRI) that is mainly used to treat premenstrual dysphoric disorder (PMDD), major depressive disorder, panic disorder, obsessive-compulsive disorder (OCD), and bulimia nervosa. It works by raising serotonin levels in the brain, which improves emotional stability and mood.

Fact

A breakthrough in the treatment of depression was made in 1987 when the U.S. Food and Drug Administration (FDA) authorized fluoxetine, the first SSRI.

Mechanism of Action

Serotonin levels in the synaptic cleft rise because fluoxetine inhibits serotonin's absorption into presynaptic nerve terminals. This increase in serotonergic activity is thought to influence fluoxetine's anxiolytic and depressive properties.

Clinical Data on Effectiveness

Clinical trials have demonstrated fluoxetine's efficacy in treating various conditions:

Major Depressive Disorder (MDD): Adult outpatients with MDD participated in a 5-week, double-blind, placebo-controlled study in which fluoxetine significantly reduced depression symptoms as assessed by the Hamilton Depression Rating Scale (HAM-D) as compared to a placebo. The effectiveness of fluoxetine in reducing depressive symptoms was demonstrated by the larger decreases in HAM-D scores observed in patients receiving the medication.

Obsessive-Compulsive Disorder (OCD): The effectiveness of fluoxetine in treating adult OCD patients was evaluated in a 13-week double-blind, placebo-controlled research. Comparing fluoxetine-treated and placebo-treated participants, the former showed a substantial decrease in OCD symptoms as assessed by the Yale-Brown Obsessive Compulsive Scale (Y-BOCS). This research validates the use of fluoxetine in treating OCD symptoms.

Bulimia Nervosa: A dose of 60 mg/day of fluoxetine significantly decreased the frequency of binge-eating and vomiting episodes when compared to a placebo in an 8-week, multicenter, double-blind, placebo-controlled study involving 387 women with bulimia nervosa diagnoses. Improvements were also observed in related attitudes and behaviors, including despair and cravings for carbohydrates. Benefits were also seen with the 20 mg/day dose, although not as much.

Panic Disorder: A 12-week, double-blind, placebo-controlled study assessed how well fluoxetine worked for people with panic disorder. The findings demonstrated fluoxetine's therapeutic potential in treating the symptoms of panic disorder by showing that patients treated with the medication had a much lower frequency of panic attacks than those receiving a placebo.

Dosage Forms Available

Fluoxetine is available in various oral forms:

  • Capsules: 10 mg, 20 mg, 40 mg, and 90 mg delayed-release.
  • Tablets: 10 mg, 20 mg, and 60 mg.
  • Oral Solution: 20 mg/5 mL.

Other Clinical Benefits

Fluoxetine has been used off-label for conditions other than its primary indications, including:

  • Premature Ejaculation: Some males may experience delayed ejaculation as a result of fluoxetine's impact on serotonin levels.
  • Fibromyalgia: Based on a few studies, fluoxetine may help reduce fibromyalgia symptoms including weariness and pain.

Side Effects Associated

Nausea, headaches, sleeplessness, dry mouth, and sexual dysfunction are typical adverse effects. Serotonin syndrome is one of the more serious side effects that can occur, particularly when paired with other serotonergic drugs.

Administration Dose and Guidelines

Each condition has a different recommended first dosage:

Depression and OCD: Usually 20 mg per day, with possible increases contingent on response.

Panic Disorder: 10 mg per day initially, then 20 mg after a week.

Individual response and tolerability should be taken into consideration while adjusting dosages.

Contraindications

Fluoxetine is contraindicated in patients:

  • Taking monoamine oxidase inhibitors (MAOIs) or within 14 days of discontinuing MAOI therapy.
  • Using pimozide or thioridazine due to the risk of QT prolongation.
  • With known hypersensitivity to fluoxetine or any of its components.
  1. Sertraline (Zoloft)

Known by the brand name Zoloft, sertraline is a selective serotonin reuptake inhibitor (SSRI) that is frequently prescribed to treat several mental illnesses, such as premenstrual dysphoric disorder, major depressive disorder (MDD), panic disorder, obsessive-compulsive disorder (OCD), and social anxiety disorder.

Fact

Sertraline was one of the most commonly prescribed antidepressants in the US as of 2022, indicating that it is widely used in clinical settings.

Mechanism of Action

Sertraline works by preventing serotonin, a neurotransmitter, from being reabsorbed into presynaptic neurons. Its antidepressant and anxiolytic effects are thought to be attributed to this inhibition, which raises serotonin availability in the synaptic cleft and improves serotonergic neurotransmission.

Clinical Data on Effectiveness

Major Depressive Disorder (MDD): In treating moderate to severe MDD, sertraline was more effective than a placebo in a multicenter, double-blind, placebo-controlled study. The Hamilton Depression Rating Scale (HDRS) showed a significant decrease in depressive symptoms over 12 weeks.

Social Anxiety Disorder (SAD): Sertraline-treated individuals showed a 40% increase in Liebowitz Social Anxiety Scale (LSAS) ratings above placebo in a 20-week double-blind study.

Dosage Forms Available

Sertraline is available in the following oral formulations:

Tablets: 25 mg, 50 mg, and 100 mg.

Oral Concentrate Solution: 20 mg/mL.

Other Clinical Benefits

Sertraline has been used off-label for illnesses like PTSD and generalized anxiety disorder in addition to its primary uses. Nevertheless, its effectiveness in treating PTSD has only demonstrated slight advantages, and more study is necessary.

Side Effects Associated

Sertraline frequently causes nausea, diarrhea, headaches, sleeplessness, dry mouth, and sexual dysfunction as adverse effects. Usually minor, these adverse effects go away with sustained use. Sertraline does, however, have a black box warning for raising the risk of suicidal thoughts and actions in kids, teens, and young adults, especially in the early stages of treatment.

Administration Dose and Guidelines

The recommended starting doses for sertraline vary by condition:

  • Major Depressive Disorder and OCD: 50 mg once daily.
  • Panic Disorder, PTSD, and Social Anxiety Disorder: 25 mg once daily, increasing to 50 mg after one week.

The highest suggested dose is 200 mg per day, and dosages can be changed in increments of 25 mg every week for at least one week. Sertraline can be taken with or without food.

Contraindications

Patients should not take sertraline:

  • Due to the possibility of serotonin syndrome, while using monoamine oxidase inhibitors (MAOIs) or within 14 days of stopping MAOI therapy.
  • In the case of co-administration of pimozide may result in severe cardiac arrhythmias.
  • If they have a history of sertraline or any of its constituents-related hypersensitivity.
  • Sertraline oral solution, which includes alcohol and may result in a disulfiram-alcohol interaction, should be taken with disulfiram.

4. GABAergic Neuromodulators

Compounds known as gamma-aminobutyric acid (GABA)ergic neuromodulators affect the action of GABA, the main inhibitory neurotransmitter in the brain. GABA regulates mood, cognition, and motor control by preserving the equilibrium between neuronal excitation and inhibition. To treat a variety of neurological and psychiatric conditions, including anxiety, epilepsy, insomnia, and alcoholism, neuromodulators that target the GABAergic system are crucial since they can either increase or decrease GABAergic transmission. These substances function by changing GABA production and breakdown, modifying receptor activation, or interacting with GABA receptors. Because of their relevance in understanding brain function and potential therapeutic uses, GABAergic neuromodulators are a focus of research.

  1. Gabapentin (Neurontin)

The U.S. Food and Drug Administration (FDA) authorized gabapentin, an anticonvulsant drug, in 1993. It is sold under the brand name Neurontin. It was first created as an anti-spasmodic and muscle relaxant, but it was later discovered to be useful as an adjuvant treatment for partial seizures and neuropathic pain.

Fact

Gabapentin's calming and euphoric effects at high doses have led to reports of misuse, especially among people with a history of substance abuse.

Mechanism of Action

The exact process by which gabapentin works is not entirely understood. By binding to the α2δ subunit of voltage-gated calcium channels in the central nervous system, it is believed to decrease excitatory neurotransmitter release, which in turn reduces pain transmission and neuronal excitability.

Clinical Data on Effectiveness

The effectiveness of gabapentin in treating neuropathic pain has been assessed in several clinical trials. According to a comprehensive review and meta-analysis of randomized controlled trials, gabapentin was more effective than a placebo, reducing pain intensity in patients with neuropathic pain by at least 30% and 50%, respectively. However, several published trials have altered primary outcomes to favor gabapentin, and several industry-sponsored trials have been challenged for result reporting bias. Furthermore, research that examined unpublished studies raised concerns about the underreporting of gabapentin side effects.

Dosage Forms Available

Gabapentin comes in a range of oral forms, such as pills, capsules, and oral solutions, and its dosages range from 100 mg to 800 mg.

Other Clinical Benefits

In addition to its core uses, gabapentin has been used to treat moderate-to-severe restless legs syndrome and neuropathic pain diseases including diabetic neuropathy.

Side Effects Associated

Gabapentin frequently causes peripheral edema, dizziness, sleepiness, and abnormalities in gait. Respiratory depression is one of the less frequent but potentially serious side effects, especially when taken with other drugs that depress the central nervous system.

Administration Dose and Guidelines

The condition being treated determines the gabapentin dosage. The recommended daily dosage for people with partial seizures is between 900 and 1800 mg, split into three doses. As tolerated, the first dosage for postherpetic neuralgia is usually 300 mg on day one, 600 mg on day two, and 900 mg on day three. Patients with renal impairment require dose modifications.

Contraindications

People who have a history of recognized hypersensitivity to gabapentin or any of its ingredients should not use it. Patients with renal impairment should use caution because their medication clearance is impaired.

  1. Diazepam (Valium)

The U.S. Food and Drug Administration (FDA) has approved the benzodiazepine drug diazepam, which is sold under the brand name Valium, for the treatment of anxiety disorders, the short-term alleviation of anxiety symptoms, spasticity related to disorders of the upper motor neurons, muscle spasms, preoperative anxiety, the treatment of some patients with refractory epilepsy, and as an adjunct in severe recurrent convulsive seizures and status epilepticus.

Fact

Compared to shorter-acting benzodiazepines, diazepam has a longer half-life, which results in longer-lasting effects and a decreased chance of withdrawal symptoms.

Mechanism of Action

The gamma-aminobutyric acid (GABA) receptor complex is where diazepam binds to increase the inhibitory effects of GABA neurotransmission. Increased neuronal hyperpolarization as a result of this interaction produces sedative, anxiolytic, anticonvulsant, and muscle relaxant effects.

Clinical Data on Effectiveness

Anxiety Disorders: In 2007, the Journal of Psychopharmacology published a systematic review and meta-analysis that assessed the effectiveness of benzodiazepines, such as diazepam, in the treatment of generalized anxiety disorder (GAD). According to the study, benzodiazepines dramatically reduced anxiety symptoms more effectively than a placebo.

Status Epilepticus: A 2000 study comparing lorazepam and diazepam for the treatment of pediatric status epilepticus was published in the New England Journal of Medicine. With a higher incidence of seizure cessation within 10 minutes of treatment, the data showed that lorazepam was more effective than diazepam at stopping seizures.

Dosage Forms Available

Diazepam is available in several formulations:

Oral Tablets: Commonly prescribed for anxiety and muscle spasm management.

Injectable Solution: Used for acute seizure management and preoperative sedation.

Rectal Gel: Indicated for the acute treatment of seizures in patients with epilepsy.

Other Clinical Benefits

Diazepam has been used for purposes outside of its primary indications, including managing the symptoms of alcohol withdrawal by lowering the chance of withdrawal seizures and giving sedation.

Side Effects Associated

Muscle weakness, exhaustion, and drowsiness are common diazepam adverse effects. Respiratory depression is one of the serious side effects that might occur, particularly when taken with other drugs that depress the central nervous system.

Administration Dose and Guidelines

The medical condition being treated determines the diazepam dosage:

Anxiety Disorders: 2-10 mg orally every 6-12 hours as needed.

Muscle Spasms: 2-10 mg orally every 3-4 hours as needed.

Status Epilepticus: 5-10 mg intravenously, repeated every 10-15 minutes if necessary, up to a total of 30 mg.

Contraindications

Patients with acute narrow-angle glaucoma, severe respiratory insufficiency, or known medication allergy should not take diazepam. Patients with a history of substance misuse, depression, or suicidal ideation should be treated with caution.

5. Glutamatergic & NMDA Modulators

Compounds that affect the glutamatergic system, the brain's main excitatory neurotransmitter network, include glutamatergic and NMDA modulators. The N-Methyl-D-Aspartate (NMDA) receptors, which are essential for synaptic plasticity, learning, and memory, are the main target of these modulators. Numerous neurological and psychiatric conditions, such as schizophrenia, depression, and Alzheimer's disease, have been linked to dysregulation of NMDA receptor function. These modulators have therapeutic potential for treating mood and cognitive problems, as well as neurodegenerative diseases, by either increasing or decreasing NMDA receptor function. Agonists, antagonists, and allosteric regulators are examples of NMDA receptor modulators that adjust receptor activity. Excitotoxicity, a process connected to neuronal damage in diseases including stroke and traumatic brain injury, can result from overactivation of NMDA receptors. On the other hand, mental symptoms and cognitive deficits are linked to decreased NMDA activation. Different kinds of NMDA modulators are exemplified by medications like ketamine, memantine, and D-cycloserine, which have uses ranging from neuroprotection and anesthesia to the treatment of depression. Novel chemicals that target NMDA receptors are being investigated in ongoing research to create safer and more effective treatments for a variety of neurological and mental health conditions.

  1. Memantine (Namenda)

Memantine is a prescription drug that is mostly used to treat moderate to severe Alzheimer's disease. It is sold under the Namenda brand. It is a member of the group of medications called NMDA (N-methyl-D-aspartate) receptor antagonists. Memantine is frequently used to enhance cognitive performance and decrease the advancement of Alzheimer's symptoms, either by itself or in conjunction with cholinesterase inhibitors such as donepezil. It can assist preserve memory, consciousness, and the capacity to carry out everyday tasks for an extended amount of time, even though it does not treat the illness.

Fact

Germany authorized memantine for medical use in 1989, and the US followed suit in 2003.

Mechanism of Action

Memantine is an uncompetitive (open-channel) NMDA receptor antagonist with low to moderate affinity. It inhibits the effects of excess glutamate, which is believed to be a contributing factor in the symptoms of Alzheimer's disease, by attaching itself to cation channels that are regulated by NMDA receptors. Excitotoxicity, a process that can cause harm to neurons, is lessened by this activity.

Clinical Data on Effectiveness

A great deal of clinical research has been conducted on memantine to assess its safety and effectiveness in treating moderate to severe Alzheimer's disease (AD). Its effects have been examined in several phase III trials that are randomized and placebo-controlled. A meta-analysis of six trials with 1,826 patients evaluated memantine's effects on behavioral symptoms, everyday activities, and cognitive function. According to the results, memantine treatment led to less decline in these areas than placebo, indicating a slight positive impact.

Dosage Forms Available

Memantine is available in several formulations:

  • Tablets: 5 mg and 10 mg strengths.
  • Extended-Release Capsules: 7 mg, 14 mg, 21 mg, and 28 mg strengths.
  • Oral Solution: 2 mg/mL concentration.

Other Clinical Benefits

Memantine has been investigated for possible advantages in various neurological conditions marked by excitotoxicity, such as vascular dementia and specific neurodegenerative disorders, in addition to its principal usage in Alzheimer's disease. However, more research is needed to determine its effectiveness in these settings.

Side Effects Associated

Constipation, headaches, dizziness, and confusion are typical memantine side effects. Hypertension, drowsiness, or hallucinations are less common side effects.

Administration Dose and Guidelines

Memantine is usually started at a dose of 5 mg once daily and increased by 5 mg per week until the target dose of 20 mg per day is reached, which is given as 10 mg twice daily. Starting at 7 mg once a day, the extended-release formulation's dosage is increased by 7 mg per week, up to a maximum of 28 mg once daily. Patients with significant renal impairment may require dose modifications.

Contraindications

Patients who have a history of known hypersensitivity to memantine or any of its ingredients should not take it. Since the pharmacokinetics of memantine have not been assessed in this population, care should be used while giving the medication to individuals who have severe hepatic impairment.

  1. Esketamine (Spravato)

Esketamine, the S-enantiomer of ketamine, is marketed as a nasal spray under the name Spravato. The U.S. Food and Drug Administration (FDA) has approved it for the treatment of major depressive disorder (MDD) and treatment-resistant depression (TRD) in patients who have acute suicidal thoughts or actions. For people who have not reacted to standard treatments, Spravato is a game-changer because it acts quickly—often within hours—in contrast to typical antidepressants, which might take weeks to start working. Spravato is only given under medical supervision in a licensed healthcare facility as part of a Risk Evaluation and Mitigation Strategy (REMS) program because of the possibility of dissociation and abuse. In addition to taking Spravato, patients also continue to take an oral antidepressant.

Fact

As the first NMDA receptor antagonist for the treatment of depression, Spravato was licensed by the U.S. Food and Drug Administration (FDA) for TRD in 2019.

Mechanism of Action

Esketamine is an ionotropic glutamate receptor antagonist that is neither selective nor competitive with the N-methyl-D-aspartate (NMDA) receptor. There is no known exact mechanism by which ketamine works as an antidepressant. AMPA receptor activation and NMDA receptor modulation are two suggested ways.

Clinical Data on Effectiveness

The effectiveness of Spravato (esketamine) nasal spray in treating treatment-resistant depression (TRD) has been assessed in several clinical trials. Patients who received Spravato plus an oral antidepressant showed a statistically significant improvement in their depression symptoms as compared to those who received a placebo plus an oral antidepressant in a key Phase 3 research. In particular, 52.5% of patients experienced remission with Spravato at Week 4, while 31.0% did so with a placebo. Furthermore, patients who received esketamine nasal spray had a 1.54-fold higher chance of achieving a therapeutic response as compared to those who received quetiapine for TRD.

Dosage Forms Available

The nasal spray Spravato contains a total of 28 mg of esketamine and comes in a device that delivers two sprays.

Other Clinical Benefits

Beyond its primary indication for TRD, esketamine has shown potential benefits in rapidly reducing depressive symptoms in patients with MDD and acute suicidal ideation or behavior.

Side Effects Associated

Dissociation, lightheadedness, nausea, drowsiness, vertigo, hypoesthesia, anxiety, sluggishness, elevated blood pressure, vomiting, and a sense of intoxication are typical adverse effects.

Administration Dose and Guidelines

An induction phase with twice-weekly administration for the first four weeks, followed by a maintenance phase with weekly or bi-weekly dosing as necessary, is the suggested dosage plan for Spravato in TRD. Because of the possibility of adverse consequences, each session needs to be monitored.

Contraindications

Patients who have a history of intracerebral hemorrhage, arteriovenous malformation, aneurysmal vascular disease, or hypersensitivity to esketamine or any of its excipients should not take Spravato.

6. Noradrenergic & Stimulant Neuromodulators

Compounds known as noradrenergic and stimulant neuromodulators affect the norepinephrine (noradrenaline) system and other neurotransmitters to control arousal, attention, and cognitive performance. The locus coeruleus-noradrenergic system, which is important for stress reactions, mood regulation, and executive function, is the main target of these modulators. Because they increase dopamine and norepinephrine activity, stimulants like methylphenidate and amphetamines are useful in the treatment of conditions including narcolepsy and attention-deficit hyperactivity disorder (ADHD). Atomoxetine and reboxetine are examples of noradrenergic drugs that specifically raise norepinephrine levels to enhance focus and impulsive control. The potential uses of these neuromodulators in neurological and psychiatric disorders, such as depression, cognitive decline, and neurodegenerative illnesses, are being extensively researched.

  1. Atomoxetine (Strattera)

Atomoxetine, a non-stimulant drug licensed for the treatment of attention-deficit/hyperactivity disorder (ADHD) in children, adolescents, and adults, is sold under the trade name Strattera. Atomoxetine, which is categorized as a selective norepinephrine reuptake inhibitor (NRI) in contrast to conventional stimulant drugs, provides an alternative for people who might not react well to stimulants or who are worried about their potential for addiction.

Fact

The U.S. Food and Drug Administration (FDA) approved atomoxetine as the first non-stimulant drug for the treatment of ADHD, giving patients an alternative therapy option.

Mechanism of Action

By specifically inhibiting the presynaptic norepinephrine transporter (NET), atomoxetine stops norepinephrine from being reabsorbed throughout the brain. This enhances adrenergic neurotransmission by raising norepinephrine levels in the synaptic cleft. Furthermore, atomoxetine indirectly raises dopamine levels in parts of the brain linked to attention and executive function, such as the prefrontal cortex.

Clinical Data on Effectiveness

Atomoxetine has been shown in clinical trials to be effective in lowering symptoms of ADHD in a range of age groups. When compared to a placebo, atomoxetine significantly improved symptomatology and functional outcomes in people with ADHD, according to a meta-analysis of six short-term studies. According to a different study, atomoxetine treatment showed small impact sizes at 4 weeks and moderate effect sizes at 6 months, suggesting that it became more effective over time.

Dosage Forms Available

Atomoxetine comes in several strengths in pill form, enabling flexible dosage that can be customized to meet the needs of each patient. Usually, the capsules are taken once or twice a day, either with or without meals.

Other Clinical Benefits

Atomoxetine has also been investigated for additional disorders in addition to its principal application in ADHD. In people with ADHD and comorbid depression, for example, it was found to dramatically reduce ADHD symptoms but did not affect depression scores.

Side Effects Associated

Atomoxetine frequently causes dry mouth, nausea, dizziness, sleeplessness, and decreased appetite as adverse effects. Blood pressure and heart rate may also rise in certain cases. Liver damage and an elevated risk of suicidal thoughts are uncommon but dangerous side effects, especially in kids and teenagers. To control these possible dangers, healthcare professionals should do routine monitoring.

Administration Dose and Guidelines

For children and adolescents up to 70 kg, the recommended first dose of atomoxetine is roughly 0.5 mg/kg; this can be raised to a goal dose of 1.2 mg/kg/day after at least three days. The starting dose for adults and individuals weighing more than 70 kg is 40 mg per day; after at least three days, the target dose can be raised to 80 mg per day. A daily intake of no more than 100 mg is advised. Clinical response and tolerability should be taken into consideration while adjusting dosage.

Contraindications

Patients with pheochromocytoma, narrow-angle glaucoma, or serious cardiovascular conditions that could be made worse by elevated blood pressure or heart rate should not use atomoxetine. Patients who have shown signs of allergy to the medication or any of its ingredients shouldn't take it. Furthermore, because atomoxetine increases the risk of hypertensive crisis, it should not be taken within 14 days of stopping an MAOI or concurrently with one.

  1. Methylphenidate (Ritalin, Concerta)

A stimulant of the central nervous system, methylphenidate is frequently given to treat narcolepsy and attention-deficit/hyperactivity disorder (ADHD). It comes in a variety of brand names, such as Ritalin and Concerta, and each one offers a unique formulation to meet the demands of the patient.

Fact

Worldwide, methylphenidate is utilized, albeit prescription rates differ from nation to nation. As ADHD has become more widely recognized and diagnosed, the number of prescriptions for it has increased in recent years.

Mechanism of Action

Methylphenidate primarily exerts its therapeutic effects by inhibiting the reuptake of dopamine and norepinephrine into presynaptic neurons. This inhibition increases the availability of these neurotransmitters in the synaptic cleft, enhancing neurotransmission and leading to improved attention and focus in individuals with ADHD.

Clinical Data on Effectiveness

Numerous studies have demonstrated the efficacy of methylphenidate in managing ADHD symptoms. A large, double-blind, randomized clinical trial demonstrated significant improvements in attention and behavior among adults with ADHD treated with methylphenidate. A meta-analysis of randomized clinical trials indicated that methylphenidate effectively reduces core ADHD symptoms in children and adolescents. However, the same analysis noted that while symptoms improved, there was limited evidence of significant enhancement in academic performance. Additionally, a multisite controlled study focusing on adolescents found that once-daily OROS methylphenidate significantly reduced ADHD symptoms and was well tolerated at dosages up to 72 mg/day.

Dosage Forms Available

Methylphenidate is available in various dosage forms:

  • Immediate-Release Tablets: Offer a quick start to action, but several dosages are needed throughout the day.
  • Extended-Release Capsules/Tablets: Intended for once-daily dosage, providing a long-lasting therapeutic impact.
  • Transdermal Patches: Deliver the medication through the skin over an extended period.

Other Clinical Benefits

Methylphenidate is authorized to treat narcolepsy, a sleep condition marked by excessive daytime sleepiness, in addition to ADHD. Its stimulant qualities encourage arousal in those who are impacted.

Side Effects Associated

Methylphenidate frequently causes the following side effects:

  • Sleeplessness
  • Diminished appetite
  • A headache
  • Pain in the abdomen
  • Elevated heart rate
  • Rarely, patients may have high blood pressure, dizziness, or mood swings.

Administration Dose and Guidelines

Methylphenidate dosage varies according to patient-specific characteristics and formulation:

  • Immediate-Release: Usually started at a low dose and titrated gradually according to tolerability and clinical response.
  • Extended-Release: Often begins with a set dose and is modified as necessary.

Contraindications

Methylphenidate is contraindicated in individuals with:

  • People who have a history of known hypersensitivity to methylphenidate or any of its ingredients
  • Significant tenseness, agitation, or anxiety
  • Glaucoma tics or a history of Tourette syndrome in the family

Clinical Applications of Neuromodulators

A variety of therapeutic approaches that alter nervous system activity are referred to as neuromodulation. These treatments are used to treat a range of neurological and psychiatric conditions as well as to support cognitive and physical rehabilitation. To change brain activity, these interventions may use pharmacological agents, electrical stimulation, or other modalities.

1. Neurological Disorders

Parkinson’s Disease

One proven neuromodulatory treatment for Parkinson's disease is deep brain stimulation (DBS). Bradykinesia, stiffness, and tremors are among the motor symptoms that DBS can reduce by sending electrical impulses to particular parts of the brain. It has been demonstrated that this method enhances the quality of life for individuals with advanced Parkinson's disease.

Epilepsy

Neuromodulation methods such as Vagus Nerve Stimulation (VNS) and Responsive Neurostimulation (RNS) have been used to lower seizure frequency in patients with drug-resistant epilepsy. For people who don't react well to pharmaceutical therapies, these devices provide therapeutic benefits by altering the brain circuits responsible for seizure genesis.

Chronic Pain

One neuromodulatory treatment for chronic pain problems is spinal cord stimulation (SCS). SCS can change how pain signals are transmitted by sending electrical pulses to the spinal cord, which helps patients with problems including failed back surgery syndrome and complicated regional pain syndrome.

2. Psychiatric Disorders

Depression and Anxiety

For depression that is resistant to treatment, repetitive transcranial magnetic stimulation (rTMS) is a non-invasive neuromodulation method that has been approved. rTMS can reduce symptoms by modifying neural activity linked to mood control by applying magnetic fields to particular brain areas.

Obsessive-Compulsive Disorder (OCD)

For severe, unresponsive OCD, deep brain stimulation (DBS) has been investigated as a potential treatment. DBS can lessen the severity of symptoms in some individuals by focusing on the brain circuits linked to obsessive-compulsive behaviors.

Addiction Treatment

The potential of neuromodulation techniques, such as rTMS and transcranial direct current stimulation (tDCS), to alter the brain circuits implicated in addiction is being studied. These methods may lessen cravings and relapse rates, according to preliminary data, but more studies are required to confirm their effectiveness.

3. Cognitive and Motor Rehabilitation

Stroke Recovery

Transcranial Direct Current Stimulation (tDCS) is one neuromodulation approach that has been used to improve neuroplasticity and functional recovery following a stroke. tDCS can improve outcomes for stroke survivors by promoting motor and cognitive rehabilitation through cortical excitability modulation.

Spinal Cord Injury Therapy

For those with spinal cord injuries, epidural spinal cord stimulation (SCS) has demonstrated promise in regaining motor function. This method can help with motor rehabilitation by activating neuronal pathways beneath the site of injury by providing electrical stimulation to the spinal cord.

Experimental and Emerging Technologies of Neuromodulation

Utilizing state-of-the-art technologies, neuromodulation is a quickly developing area that modifies brain activity for both therapeutic and enhancing objectives. Neuromodulation has historically depended on electrical stimulation methods including spinal cord stimulation (SCS) and deep brain stimulation (DBS). Closed-loop neuromodulation, optogenetics, gene therapy, brain-computer interfaces (BCIs), and artificial intelligence (AI)-driven neural modulation are some of the more advanced and accurate methods that have been made possible by recent developments. The goal of these cutting-edge and experimental technologies is to treat neurological conditions like epilepsy, Parkinson's disease, depression, and chronic pain in a highly customized, adaptable, and non-invasive manner. The next generation of neuromodulation technologies is opening the door to safer, more efficient, and patient-specific therapies by combining machine learning algorithms, real-time feedback systems, and minimally invasive procedures.

1. Closed-Loop Neuromodulation

Closed-loop neuromodulation is a significant advancement in neurotechnology that offers a dynamic and personalized way to stimulate the nervous system and brain. Unlike traditional open-loop systems that deliver preset stimulation regardless of a patient's real-time neural activity, closed-loop systems continuously analyze brain signals and adjust stimulation parameters in response to them. Because it improves efficacy while reducing side effects, this real-time feedback system holds particular promise for treating neurological disorders like epilepsy, Parkinson's disease, chronic pain, and depression.

How It Works?

Closed-loop neuromodulation consists of a three-step process:

Real-Time Monitoring Systems

Sensors integrated into brain implants or wearable devices measure electrical activity, neurotransmitter levels, and other physiological markers. These devices monitor neural impulses and adjust stimulation in response to bodily changes. For instance, epilepsy is treated using devices that can detect the beginning of seizures and stimulate the brain quickly to prevent further progression. One such example is the Picostim implant, which has shown promising results in reducing seizures.

Data Processing & Decision-Making

Advanced algorithms, frequently driven by artificial intelligence (AI) or machine learning, are used to analyze the gathered brain signals in order to find anomalies or patterns suggestive of disease symptoms.

Adaptive Stimulation Techniques

The system modifies stimulation duration, frequency, or strength based on real-time data to maximize therapeutic benefits and reduce side effects. Adaptive systems increase effectiveness and reduce adverse effects by adjusting stimulation parameters in response to continuous brain activity. Multiphoton microscopy research has made closed-loop studies possible, enabling real-time brain circuit observation and modification.

Applications of Closed-Loop Neuromodulation

  • Epilepsy Treatment: Seizures can be avoided or reduced with the use of devices such as the NeuroPace RNS System, which identify aberrant brain activity before a seizure occurs. According to studies, this technology can help many patients experience fewer seizures by more than 70%.
  • Parkinson’s Disease Management: Closed-loop deep brain stimulation (DBS) devices, like those being tested in clinical trials at UCSF, modify stimulation in response to brain signals related to movement, minimizing motor fluctuations and tremors while conserving energy.
  • Chronic Pain Therapy: Spinal cord stimulators, like Medtronic's Evoke System, evaluate brain responses to pain constantly and adjust stimulation in real time to improve alleviation while using less power.
  • Mental Health Applications: Personalized DBS, which uses real-time monitoring of mood-related brain activity to guide stimulation modifications for more effective symptom relief, is being investigated as an adaptive neuromodulation treatment for treatment-resistant depression (TRD).

Future Prospects

AI-driven decision-making, wireless implantable sensors, and multi-modal stimulation—which combines electrical, optical, and pharmaceutical interventions—are key components of closed-loop neuromodulation's future. Device longevity and patient comfort will be further improved by developments in biocompatible materials and miniaturization. Closed-loop systems are anticipated to become a common therapeutic strategy as research advances, providing precision medicine for a variety of neurological and psychiatric conditions.

2. Optogenetics and Gene Therapy

Neuromodulation is being revolutionized by optogenetics and gene therapy, which allow for precise, cell-specific regulation of brain circuits. By employing light-sensitive proteins, optogenetics enables the targeted activation or inhibition of particular neuronal subtypes, in contrast to conventional electrical stimulation, which impacts vast populations of neurons. By delivering genetic alterations to improve or restore neural function, gene therapy enhances this strategy and provides prospective therapies for neurological conditions with genetic roots. When combined, these state-of-the-art technologies have the potential to cure illnesses including epilepsy, spinal cord injury, Parkinson's disease, and even neuropsychiatric problems.

How It Works?

Optogenetics Mechanism

Opsins, which are light-sensitive proteins, are introduced into specific neurons via gene therapy. Certain light wavelengths given by fiber-optic implants or non-invasive methods like transcranial optogenetic stimulation can be used to either activate or inhibit these neurons once they have been produced.

Gene Therapy Mechanism

Therapeutic genes can be introduced into the nervous system by viral vectors like adeno-associated viruses (AAVs) in order to alter neurotransmitter release, encourage neuronal regeneration, or fix genetic abnormalities.

Precision Neuromodulation

Optogenetics and gene therapy minimize adverse effects by precisely controlling brain activity with millisecond resolution, in contrast to older approaches that lack cellular specificity.

Applications of Optogenetics and Gene Therapy

  • Parkinson’s Disease Treatment: Compared to conventional deep brain stimulation (DBS), optogenetic stimulation of the subthalamic nucleus (STN) has been demonstrated to restore motor function more precisely while minimizing undesired side effects. Furthermore, by increasing GABA synthesis to rebalance brain circuits, gene therapy techniques like AAV-GAD (glutamic acid decarboxylase) therapy have shown long-term advantages in clinical trials.
  • Epilepsy Control: In animal models, optogenetics has been utilized to turn off hyperactive neurons as seizures start, which could result in a next-generation substitute for implanted electrical stimulators such as the NeuroPace RNS System.
  • Spinal Cord Injury and Paralysis Recovery: While optogenetics allows for precise control over motor neuron activity to restore movement in paralyzed persons, gene therapy-based strategies, such as the introduction of chondroitinase ABC (ChABC), encourage axonal regeneration.
  • Depression and Anxiety Disorders: Understanding the neuronal circuits underpinning mood regulation has been made possible by optogenetic stimulation of the amygdala and prefrontal cortex. Non-invasive optogenetics may be used in future therapies to reduce the symptoms of PTSD and treatment-resistant depression (TRD).

Future Prospects

Non-invasive light delivery methods, genetically modified opsins with increased sensitivity, and integration with artificial intelligence (AI) for adaptive neuromodulation are the next frontiers in optogenetics and gene therapy. Invasive optical fibers may become unnecessary with the development of magnetically controlled optogenetics and ultrasound-triggered gene activation. As these technologies develop, they will push the limits of precision medicine in neuromodulation by enabling highly focused and durable treatments for a variety of neurological and psychiatric diseases.

3. Brain-Computer Interfaces (BCI) and AI Integration

By facilitating smooth communication between the brain and external devices, artificial intelligence (AI) and brain-computer interfaces (BCIs) are revolutionizing neuromodulation. By converting neural activity into digital signals, brain-computer interfaces (BCIs) enable people to operate computers, prosthetics, or neuromodulatory implants with just their thoughts. By deciphering intricate brain patterns, customizing stimulation regimens, and instantly improving therapeutic results, the incorporation of AI significantly improves these devices. Novel therapies for ailments like paralysis, epilepsy, depression, and neurodegenerative diseases are being made possible by this combination of neuroscience and machine learning.

How It Works?

Neural Signal Acquisition

BCIs monitor electrical activity from the brain using electrodes, either non-invasive or implanted, usually using intracortical arrays, electroencephalography (EEG), or electrocorticography (ECoG).

AI-Powered Signal Processing

These signals are decoded by sophisticated machine learning algorithms, which find patterns connected to defects caused by sickness, motor intent, or cognitive states.

Adaptive Neuromodulation

AI-powered closed-loop systems make real-time adjustments to neuromodulation parameters, guaranteeing accurate stimulation to improve cognitive function, alleviate symptoms, or restore lost functions.

Applications of BCI and AI

  • Restoring Motor Function in Paralysis: For people with spinal cord injuries, brain-controlled robotic limbs and exoskeletons are now possible thanks to BCIs. By more accurately anticipating movement intentions, artificial intelligence (AI) enhances these systems and makes prosthetic devices more responsive. Patients who are paralyzed can now operate digital devices with just their thoughts thanks to Elon Musk's Neuralink.
  • Epilepsy and Seizure Prediction: Artificial intelligence (AI)-driven brain-computer interfaces (BCIs) are able to identify early indicators of epileptic seizures and initiate closed-loop neuromodulation devices, including responsive neurostimulation (RNS), to inhibit aberrant brain activity prior to a seizure.
  • Depression and Mood Disorders: Treatment-resistant depression (TRD) may be treated with customized AI-driven DBS devices that can recognize the neural markers of depression and provide focused stimulation to areas of the brain that control mood.
  • Cognitive Enhancement and Neuroprosthetics: Memory enhancement implants, like those created by DARPA, have been made possible by BCIs combined with AI. These implants increase recollection by activating hippocampus circuits. Alzheimer's disease and other cognitive diseases may be treated with these technology.

Future Prospects

Wireless, minimally invasive neural interfaces, non-invasive BCIs that use functional near-infrared spectroscopy (fNIRS) or optogenetics, and AI models that anticipate and adjust to unique patterns of brain activity will be the main focuses of the next generation of BCI and AI-driven neuromodulation. Future advancements in bidirectionally communicating brain implants, like Synchron's Stentrode, will further close the gap between machine intelligence and human cognition, paving the way for highly customized and successful neuromodulatory treatments.

4. Magnetothermal Stimulation

Magnetothermal stimulation is a new neuromodulation method that uses thermal energy and magnetic fields to wirelessly modulate neuronal activity. With the ability to precisely target particular brain circuits, this technique provides a less intrusive option to conventional electrical stimulation. Magnetothermal stimulation is being investigated as a potential treatment for neurological conditions such epilepsy, Parkinson's disease, and chronic pain.

How It Works?

Nanoparticle Introduction

Superparamagnetic nanoparticles are delivered systemically or by direct injection into the target brain tissue. The purpose of these nanoparticles is to react to magnetic fields outside of them.

Magnetic Field Application

External application of an alternating magnetic field (AMF) allows it to deeply penetrate biological tissues with little to no attenuation.

Localized Heating

The AMF causes the nanoparticles to oscillate quickly, producing heat locally.

Neuronal Activation

Depolarization and the start of action potentials result from the localized heating activating temperature-sensitive ion channels, such as TRPV1, which is expressed on neurons.

Applications of Magnetothermal Stimulation

  • Parkinson's Disease: Magnetothermal techniques have been shown to reduce parkinsonian-like symptoms in mice models. In two animal models of Parkinson's disease, Hescham et al. (2021) used magnetothermal nanoparticle technology to produce therapeutic results similar to those of conventional deep brain stimulation (DBS). Their findings were reported in Nature Communications.
  • Epilepsy: The use of magnetothermal stimulation to alter the brain circuits responsible for seizure activity has been investigated. Wireless magnetothermal deep brain stimulation was demonstrated in a study, laying the groundwork for possible uses in the treatment of epilepsy.
  • Chronic Pain: Magnetothermal stimulation presents a viable approach to modifying pain perception by focusing on particular pain pathways. Studies examining the effectiveness of this strategy in modifying the brain circuits linked to pain are still being conducted in this field.

Future Directions

Magnetothermal stimulation advancements are concentrated on:

  • Nanoparticle Optimization: Creating biocompatible nanoparticles that can be delivered to particular neuronal populations with improved magnetic characteristics.
  • Non-Invasive Delivery: Investigating non-invasive ways to administer magnetic fields and nanoparticles to reduce procedure risks and patient pain.
  • Integration with Other Modalities: Combining magnetothermal stimulation with other neuromodulation methods, including pharmaceutical treatments or optogenetics, to provide therapeutic results that work in concert.

5. Flexible Neural Interfaces

An important development in neuromodulation is the use of flexible neural interfaces, which are more functional and biocompatible than conventional rigid devices. These interfaces improve signal integrity and lessen tissue damage by adapting to the intricate, dynamic architecture of brain tissue. This increases the effectiveness of neuromodulatory therapy for diseases like epilepsy, Parkinson's disease, and chronic pain.

How They Work?

Material Composition

Biocompatible materials that offer the required flexibility and endurance, such as silicone, polyimide, or parylene, are used to build flexible neural interfaces.

Design Architecture

These devices can tightly adhere to neural tissues because they use novel structural designs including mesh-like arrangements or serpentine traces.

Functionality

Flexible interfaces with electrodes or optoelectronic components can offer specific electrical or optical stimulation and record neural activity.

Applications in Neuromodulation

  • Epilepsy Management: Seizures have been seen and their activity modulated using flexible neural interfaces. A flexible, biodegradable silicon-based neural interface with transdermal optoelectronic stimulation capabilities was presented in a study. This technology provides a minimally invasive method of brain modulation.
  • Parkinson's Disease Treatment:Researchers examined current developments in electrical neuromodulation implantable devices in Advanced Healthcare Materials, emphasizing the function of flexible interfaces in providing more potent treatments for movement disorders.
  • Chronic Pain Relief: In an effort to reduce chronic pain, flexible neural interfaces are being investigated for spinal cord stimulation. Their conformability makes it possible to precisely target the brain pathways that are involved in pain perception, which may improve the effectiveness of treatment.

Future Prospects

Flexible neural interfaces have the potential to be a key component of next-generation neuromodulatory therapies, which will provide individualized and efficient treatments for a variety of neurological conditions as research advances. Flexible neural interface developments are concentrating on:

  • Multimodal Integration: Integrating chemical, optical, and electrical modalities to produce multipurpose tools with complete neuromodulation capabilities.
  • Biodegradable Materials: Creating interfaces using materials that break down naturally in the body, removing the need for surgery and lowering the risk of long-term issues.
  • High-Density Electrode Arrays: Increasing the electrode density allows for more accurate mapping and neural circuit modulation while also improving spatial resolution.

6. Transcranial Focused Ultrasound (tFUS)

A new neuromodulation method called transcranial focused ultrasound (tFUS) allows for targeted, non-invasive changes in brain activity. Without requiring surgery, tFUS may precisely interact with deep brain areas by penetrating the skull with focused ultrasound waves. Numerous neurological and psychological conditions, such as depression, epilepsy, and movement disorders, may benefit from this treatment strategy.

How It Works?

Ultrasound Wave Generation

Low-intensity ultrasonic waves at particular frequencies are released by a transducer.

Skull Penetration

These waves can reach specific brain regions by non-invasively penetrating the skull.

Focal Modulation

Targeted neuromodulation is possible because the ultrasonic waves are concentrated on a tiny, exact location.

Neuronal Interaction

Therapeutic benefits may result from the concentrated ultrasound's ability to alter neuronal activity.

Applications in Neuromodulation

  • Movement Disorders: As a non-invasive substitute for conventional techniques, tFUS has been investigated as a treatment for movement disorders.
  • Epilepsy: Based on research, tFUS may be able to modify the brain circuits responsible for seizure activity, offering a non-invasive treatment option for epilepsy.
  • Depression: Research has looked into how tFUS can alter the parts of the brain linked to mood regulation, which may have uses in the treatment of depression.

Future Prospects

tFUS has great promise as a precise, non-invasive neuromodulation technique for a variety of clinical uses as research advances. tFUS advancements are concentrated on:

  • Mechanistic Understanding: To clarify the exact processes by which tFUS alters neuronal activity, more investigation is required.
  • Parameter Optimization: For clinical use, figuring out the ideal ultrasonic parameters to produce the intended neuromodulatory effects is essential.
  • Clinical Trials: Clinical trials are being conducted now and in the future to determine the safety and effectiveness of tFUS in treating a range of neurological and psychiatric conditions.

Challenges and Ethical Considerations of Neuromodulation

Patients with illnesses including Parkinson's disease, epilepsy, depression, and chronic pain now have hope thanks to neuromodulation, which has transformed the treatment of numerous neurological and psychiatric problems. But despite its potential, neuromodulation has a number of drawbacks and moral issues that need to be properly considered. These worries cover everything from the wider ethical ramifications of altering brain activity to regulatory obstacles and unexpected side effects. Thorough clinical research, ethical supervision, and legislative changes are necessary to address these problems and guarantee that neuromodulatory treatments continue to be secure, efficient, and available to all patients who require them. A multidisciplinary strategy combining neuroscientists, ethicists, legislators, and clinicians will be crucial to directing the responsible development and use of these technologies as they continue to advance.

1. Risks and Side Effects of Neuromodulation

Even while neuromodulation technologies have many therapeutic advantages, there are certain hazards associated with them, such as the possibility of surgical complications, device malfunctions, and unexpected emotional or cognitive impacts. Depending on the particular neuromodulation method employed, these hazards change.

A. Surgical Risks and Infections

  • Electrodes and stimulation devices must be implanted for invasive treatments like Deep Brain Stimulation (DBS) and Vagus Nerve Stimulation (VNS), which entail hazards like bleeding, infection, and immunological reaction.
  • Nerve injury or tissue damage may result from spinal cord stimulation (SCS), especially if electrodes move from their intended location.
  • Device failure and post-surgical infections are among the issues that 5–10% of DBS patients encounter, according to a study published in JAMA Neurology (Lozano et al., 2019).

B. Unintended Cognitive and Emotional Effects

  • Cognitive deficits, personality changes, and mood swings can occasionally result from neuromodulation. For instance, a research by Hariz et al. (2021) in The Lancet Neurology noted that some Parkinson's patients receiving DBS have reported having impulsivity, despair, or hypomania.
  • Despite being non-invasive, tDCS (Transcranial Direct Current Stimulation) and tFUS (Transcranial Focused Ultrasound) can result in headaches, lightheadedness, or unexpected cognitive disturbances if applied incorrectly.

C. Long-Term Effects and Device Malfunctions

  • Long-term implantation is necessary for many neuromodulation treatments, which raises questions about device deterioration, battery failures, and the necessity for repeated surgeries.
  • According to an FDA Medical Device Reports (MDRs) report, there have been instances of SCS and DBS devices failing too soon, requiring device explants and reimplantations.

2. Ethical Concerns in Brain Manipulation

Basic ethical precepts pertaining to autonomy, identity, and consent are called into question by neuromodulation. Concerns over how these technologies affect a person's personal agency, cognition, and emotions are raised by their capacity to modify brain activity.

A. Autonomy and Informed Consent

  • Neuromodulation patients need to be well aware of the dangers, advantages, and long-term effects of these procedures.
  • Certain mental and neurological disorders (such as obsessive-compulsive disorder and severe depression) can affect a patient's capacity to make decisions, which raises questions regarding whether or not they can actually give informed consent.
  • The moral obligation of doctors to prevent patients from being forced to undergo neuromodulation therapies is covered in a research that was published in Neuroethics (Fins et al., 2020).

B. Identity and Personality Changes

  • Changes in personality, emotions, and self-perception have been documented as a result of neuromodulation procedures, specifically DBS and closed-loop stimulation systems.
  • After receiving DBS, some Parkinson's patients have reported feeling like a new person, which raises ethical questions regarding how these therapies could change a person's fundamental identity.
  • The philosophical conundrum of whether a person who has undergone neuromodulation therapy is still the same person who first gave their consent is brought to light by Gilbert et al. (2018) in Frontiers in Human Neuroscience.

C. Potential for Cognitive Enhancement and Misuse

  • Although the main application of neuromodulation is in medicine, there is increasing interest in employing these technologies to improve cognitive function in healthy people.
  • The military has investigated tDCS to improve troops' memory and cognitive function, which raises moral concerns regarding the possibility of coercion in high-stress occupations.
  • Social inequality and unfair advantages could result from the potential for "brain hacking" or neurostimulation-based cognitive upgrades in competitive domains (sport, academia, etc.).

3. Regulatory and Accessibility Challenges

Significant financial, regulatory, and accessibility obstacles prevent neuromodulation technology from being widely used and distributed fairly.

A. Approval and Safety Regulations

The European Medicines Agency (EMA) and the U.S. Food and Drug Administration (FDA) require neuromodulation devices to pass stringent clinical trials and receive regulatory approval. However, access to potentially life-changing treatments might be delayed for years due to approval procedures for implanted devices (such as DBS and SCS). Concerns regarding uneven safety testing procedures for neuromodulatory implants were brought to light in a 2022 study by the FDA's Center for Devices and Radiological Health (CDRH).

B. Cost and Insurance Coverage

Many people cannot afford neuromodulation therapy since DBS implantation costs between $50,000 and $100,000 per patient. Due to a lack of long-term data on efficacy, insurance companies frequently restrict reimbursement for neuromodulation therapies, especially non-invasive methods like tDCS or tFUS. Only 30–40% of Parkinson's patients who could benefit from DBS actually receive it, according to a review published in The Lancet Neurology in 2021. This is primarily because to insurance and financial constraints.

C. Global Disparities in Access to Neuromodulation

Modern neuromodulation treatments are more widely available in high-income nations, but low- and middle-income nations (LMICs) have major obstacles, such as a lack of money, infrastructure, and qualified specialists. According to a 2023 World Health Organization (WHO) assessment on neuromodulation technology, international efforts are required to increase accessibility and affordability for marginalized communities.

Conclusion

From crude electrical therapies to sophisticated, individualized, and precise treatment modalities, neuromodulation has transformed and now offers hope to patients with a range of neurological and psychiatric conditions. Neuromodulation has transformed the treatment of diseases like Parkinson's disease, epilepsy, chronic pain, and depression by restoring or regulating neuronal function by electrical, chemical, and non-invasive methods.

The development of novel technologies like as gene therapy, closed-loop neuromodulation, optogenetics, brain-computer interfaces (BCIs), and AI-driven neuromodulation has improved the accuracy, versatility, and efficacy of treatment. These developments have the potential to advance the field of truly customized medicine by enhancing patient outcomes while reducing adverse effects. Neuromodulation is becoming safer and more accessible because to non-invasive methods like flexible neural interfaces and transcranial focused ultrasound (tFUS), which also lessen the need for surgical procedures.

But these developments also bring with them serious difficulties and moral dilemmas. Through thorough research and professional supervision, the risks of neuromodulation—which might range from unanticipated cognitive and emotional alterations to surgical complications—must be properly handled. Scientists, ethicists, and politicians must continue to discuss ethical conundrums, such as those involving informed consent, identity changes, and the possibility of cognitive improvement or abuse. Furthermore, universal accessibility is restricted by financial and legal obstacles, especially in areas with low and middle incomes.

A multidisciplinary strategy combining neuroscientists, clinicians, engineers, legislators, and ethicists will be crucial as neuromodulation advances to guarantee safe, efficient, and fair access to these transformative technologies. Neuromodulatory therapies will be further refined in the future, especially in the areas of biomaterial breakthroughs, wireless implanted devices, and AI integration, opening the door to more accurate, adaptable, and least invasive treatments. Neuromodulation has the potential to revolutionize neurological care and enhance the lives of millions of people worldwide with further study, ethical supervision, and regulatory developments.

REFERENCES

History of Neuromodulation.https://www.neuromodulation.com/brief-history-neuromodulation. International Neuromodulation Society, February 4, 2025.

Herrington, T.M., Cheng, J.J. and Eskandar, E.N., 2016. Mechanisms of deep brain stimulation. Journal of neurophysiology, 115(1), pp.19-38.

Sundaram, H., Signorini, M., Liew, S., de Almeida, A.R.T., Wu, Y., Braz, A.V., Fagien, S., Goodman, G.J., Monheit, G., Raspaldo, H. and Global Aesthetics Consensus Group, 2016. Global aesthetics consensus: botulinum toxin type A—evidence-based review, emerging concepts, and consensus recommendations for aesthetic use, including updates on complications. Plastic and reconstructive surgery, 137(3), pp.518e-529e.

Attal, N., de Andrade, D.C., Adam, F., Ranoux, D., Teixeira, M.J., Galhardoni, R., Raicher, I., Üçeyler, N., Sommer, C. and Bouhassira, D., 2016. Safety and efficacy of repeated injections of botulinum toxin A in peripheral neuropathic pain (BOTNEP): a randomised, double-blind, placebo-controlled trial. The Lancet Neurology, 15(6), pp.555-565.

Beer, K.R., Shamban, A.T., Avelar, R.L., Gross, J.E. and Jonker, A., 2019. Efficacy and safety of prabotulinumtoxinA for the treatment of glabellar lines in adult subjects: results from 2 identical phase III studies. Dermatologic Surgery, 45(11), pp.1381-1393.

Frequin, H.L., Schouten, J., Verschuur, C.V., Suwijn, S.R., Boel, J.A., Post, B., Bloem, B.R., van Hilten, J.J., van Laar, T., Tissingh, G. and Munts, A.G., 2023. Levodopa response in patients with early Parkinson disease: further observations of the LEAP study. Neurology, 100(4), pp.e367-e376.

Parkinson Study Group and Parkinson Study Group, 2000. Pramipexole vs levodopa as initial treatment for Parkinson disease: a randomized controlled trial. Jama, 284(15), pp.1931-1938.

Ferini-Strambi, L., Aarskog, D., Partinen, M., Chaudhuri, K.R., Sohr, M., Verri, D. and Albrecht, S., 2008. Effect of pramipexole on RLS symptoms and sleep: a randomized, double-blind, placebo-controlled trial. Sleep medicine, 9(8), pp.874-881.

Fluoxetine Bulimia Nervosa Collaborative Study Group, 1992. Fluoxetine in the treatment of bulimia nervosa. A multicenter, placebo controlled, double-blind trial. Arch. Gen. Psychiatry, 49, pp.139-147.

Beasley Jr, C.M., Koke, S.C., Nilsson, M.E. and Gonzales, J.S., 2000. Adverse events and treatment discontinuations in clinical trials of fluoxetine in major depressive disorder: an updated meta-analysis. Clinical therapeutics, 22(11), pp.1319-1330.

Geller, D.A., Hoog, S.L., Heiligenstein, J.H., Ricardi, R.K., Tamura, R.O.Y., Kluszynski, S., Jacobson, J.G. and Fluoxetine Pediatric OCD Study Team, 2001. Fluoxetine treatment for obsessive-compulsive disorder in children and adolescents: a placebo-controlled clinical trial. Journal of the American Academy of Child & Adolescent Psychiatry, 40(7), pp.773-779.

Michelson, D., Lydiard, R.B., Pollack, M.H., Tamura, R.N., Hoog, S.L., Tepner, R., Demitrack, M.A., Tollefson, G.D. and Fluoxetine Panic Disorder Study Group, 1998. Outcome assessment and clinical improvement in panic disorder: evidence from a randomized controlled trial of fluoxetine and placebo. American Journal of Psychiatry, 155(11), pp.1570-1577.

Lépine, J.P., Caillard, V., Bisserbe, J.C., Troy, S., Hotton, J.M., Boyer, P. and PREVERS Investigators, 2004. A randomized, placebo-controlled trial of sertraline for prophylactic treatment of highly recurrent major depressive disorder. American Journal of Psychiatry, 161(5), pp.836-842.

Mandrioli, R., Mercolini, L. and Raggi, M.A., 2013. Evaluation of the pharmacokinetics, safety and clinical efficacy of sertraline used to treat social anxiety. Expert opinion on drug metabolism & toxicology, 9(11), pp.1495-1505.

Meaadi, J., Obara, I., Eldabe, S. and Nazar, H., 2023. The safety and efficacy of gabapentinoids in the management of neuropathic pain: a systematic review with meta-analysis of randomised controlled trials. International Journal of Clinical Pharmacy, 45(3), pp.556-565.

Offidani, E., Guidi, J., Tomba, E. and Fava, G.A., 2013. Efficacy and tolerability of benzodiazepines versus antidepressants in anxiety disorders: a systematic review and meta-analysis. Psychotherapy and psychosomatics, 82(6), pp.355-362.

Alldredge, B.K., Gelb, A.M., Isaacs, S.M., Corry, M.D., Allen, F., Ulrich, S., Gottwald, M.D., O'Neil, N., Neuhaus, J.M., Segal, M.R. and Lowenstein, D.H., 2001. A comparison of lorazepam, diazepam, and placebo for the treatment of out-of-hospital status epilepticus. New England Journal of Medicine, 345(9), pp.631-637.

Winblad, B., Jones, R.W., Wirth, Y., Stöffler, A. and Möbius, H.J., 2007. Memantine in moderate to severe Alzheimer’s disease: a meta-analysis of randomised clinical trials. Dementia and geriatric cognitive disorders, 24(1), pp.20-27.

Reif, A., Bitter, I., Buyze, J., Cebulla, K., Frey, R., Fu, D.J., Ito, T., Kambarov, Y., Llorca, P.M., Oliveira-Maia, A.J. and Messer, T., 2023. Esketamine nasal spray versus quetiapine for treatment-resistant depression. New England Journal of Medicine, 389(14), pp.1298-1309.

Asherson, P., Bushe, C., Saylor, K., Tanaka, Y., Deberdt, W. and Upadhyaya, H., 2014. Efficacy of atomoxetine in adults with attention deficit hyperactivity disorder: an integrated analysis of the complete database of multicenter placebo-controlled trials. Journal of Psychopharmacology, 28(9), pp.837-846.

de Faria, J.C.M., Duarte, L.J.R., Ferreira, L.D.A., da Silveira, V.T., Menezes de Pádua, C. and Perini, E., 2022. “Real‐world” effectiveness of methylphenidate in improving the academic achievement of Attention‐Deficit Hyperactivity Disorder diagnosed students—A systematic review. Journal of Clinical Pharmacy and Therapeutics, 47(1), pp.6-23.

Edwards, C.A., Kouzani, A., Lee, K.H. and Ross, E.K., 2017, September. Neurostimulation devices for the treatment of neurologic disorders. In Mayo Clinic Proceedings (Vol. 92, No. 9, pp. 1427-1444). Elsevier.

Wolpaw, J.R., 2012. Harnessing neuroplasticity for clinical applications. Brain, 135(4), pp.e215-e215.

Kammen, A., Cavaleri, J., Lam, J., Frank, A.C., Mason, X., Choi, W., Penn, M., Brasfield, K., Van Noppen, B., Murray, S.B. and Lee, D.J., 2022. Neuromodulation of OCD: A review of invasive and non-invasive methods. Frontiers in neurology, 13, p.909264.

Kollewe, J. (2024). Brain implants to treat epilepsy, arthritis, or even incontinence? They may be closer than you think. [online] the Guardian. Available at: https://www.theguardian.com/business/article/2024/aug/17/brain-implants-to-treat-epilepsy-arthritis-or-even-incontinence-they-may-be-closer-than-you-think? [Accessed 9 Feb. 2025].

Hira, R., 2024. Closed-loop experiments and brain machine interfaces with multiphoton microscopy. Neurophotonics, 11(3), pp.033405-033405.

Hescham, S.A., Chiang, P.H., Gregurec, D., Moon, J., Christiansen, M.G., Jahanshahi, A., Liu, H., Rosenfeld, D., Pralle, A., Anikeeva, P. and Temel, Y., 2021. Magnetothermal nanoparticle technology alleviates parkinsonian-like symptoms in mice. Nature communications, 12(1), p.5569.

Wang, L., Liu, S., Zhao, W., Li, J., Zeng, H., Kang, S., Sheng, X., Wang, L., Fan, Y. and Yin, L., 2024. Recent Advances in Implantable Neural Interfaces for Multimodal Electrical Neuromodulation. Advanced Healthcare Materials, p.2303316.

Zhang, T., Pan, N., Wang, Y., Liu, C. and Hu, S., 2021. Transcranial focused ultrasound neuromodulation: a review of the excitatory and inhibitory effects on brain activity in human and animals. Frontiers in Human Neuroscience, 15, p.749162.

Fins, J.J. and Shulman, K.S., 2024. Neuroethics, Covert Consciousness, and Disability Rights: What Happens When Artificial Intelligence Meets Cognitive Motor Dissociation?. Journal of Cognitive Neuroscience, pp.1-8.

Gilbert, F., 2018. Deep brain stimulation: Inducing self-estrangement. Neuroethics, 11(2), pp.157-165.

Bloem, B.R., Okun, M.S. and Klein, C., 2021. Parkinson's disease. The Lancet, 397(10291), pp.2284-2303.