Fact-checked by Grok 2 weeks ago

Sodium channel blocker

Sodium channel blockers are a class of pharmacological agents that inhibit the conductance of sodium ions through voltage-gated sodium channels, which are essential transmembrane proteins in excitable cells such as neurons, cardiac myocytes, and fibers. These channels, composed primarily of a large alpha subunit forming the ion-selective pore and smaller beta subunits that regulate gating and localization, open in response to membrane depolarization to allow rapid sodium influx, initiating and propagating action potentials. By preventing this influx, sodium channel blockers reduce cellular excitability, stabilizing membrane potentials and suppressing abnormal electrical activity. The involves drug binding to specific sites within the 's inner , often in the S6 helices of domains III and , where they exert electrostatic and steric blockade of . This binding exhibits state-dependence, with higher affinity for open or inactivated conformations, leading to use-dependent or frequency-dependent inhibition that preferentially targets rapidly firing cells during pathological conditions like arrhythmias or seizures. Both charged (cationic) and neutral drugs achieve blockade, with cationic agents like lidocaine directly occupying the and neutral ones like trapping sodium to disrupt flow. Clinically, sodium channel blockers are categorized by therapeutic application and subclass, including Class I antiarrhythmics (e.g., quinidine, procainamide, lidocaine, flecainide) that slow cardiac conduction to treat ventricular and supraventricular tachydysrhythmias by decreasing phase 0 velocity. Local anesthetics such as bupivacaine and block impulses for regional , while anticonvulsants like , , and prevent propagation by limiting neuronal firing. Additional uses include management of with agents like tricyclic antidepressants (e.g., amitriptyline), which also exhibit sodium channel blockade alongside other effects. These drugs' versatility stems from their shared target but varies in , potency, and selectivity across isoforms like Nav1.5 in the heart or Nav1.7 in pain pathways.

Introduction and Mechanism

Definition and Basic Mechanism

Sodium channel blockers are pharmacological agents that inhibit the function of voltage-gated sodium () channels, preventing sodium influx across membranes. These compounds bind to specific sites on the protein, modulating its gating properties and thereby altering the excitability of cells such as neurons and cardiomyocytes. The basic mechanism of sodium channel blockers involves reducing the rate of depolarization in excitable cells by stabilizing the channels in inactivated or non-conducting states. This binding decreases the sodium conductance (gNa), which limits the influx of sodium ions during the action potential upstroke, resulting in reduced action potential amplitude and slowed conduction velocity. The sodium current (INa) can be described by the equation: I_{Na} = g_{Na} (V - E_{Na}) where V is the membrane potential and E_{Na} is the sodium equilibrium potential; blockade primarily reduces g_{Na}. Local anesthetics exemplify this by preferentially binding to the open or inactivated conformations, enhancing inactivation and preventing channel recovery. The pharmacological action of sodium channel blockers was first identified in the 1950s through studies on local anesthetics like , which were shown to suppress sodium currents in voltage-clamped axons. A key milestone came in 1964 with the discovery that (TTX), a from pufferfish, acts as a highly selective natural blocker by occluding the sodium conduction pathway. Major categories of these agents include antiarrhythmics and anesthetics, which find therapeutic applications in and .

Clinical Significance

Sodium channel blockers play a crucial role in modulating excitability across neuronal, cardiac, and muscular tissues by inhibiting voltage-gated sodium channels, thereby altering the propagation of action potentials and mitigating hyperexcitability disorders. In neuronal tissues, they suppress abnormal firing associated with conditions like and , while in , they stabilize membrane potentials to prevent erratic rhythms; in skeletal and smooth , they reduce conduction to manage local pain and spasms. These agents target diseases such as cardiac arrhythmias, including affecting 2-3% of the general population as of 2025, alongside impacting approximately 52 million people worldwide as of 2021 estimates, and prevalent in 7-10% of the general population. The of sodium channel blockers hinge on state-dependent , where they preferentially interact with inactivated or open channel states during high-frequency activity, allowing for a therapeutic window that permits partial blockade to dampen pathological excitability without inducing complete conduction block or toxicity. This selectivity ensures onset and duration are influenced by the tissue's firing rate—rapid in hyperexcitable states like arrhythmias or seizures—enabling effective symptom control while preserving normal physiological function. Such principles underpin their utility in clinical settings, where achieving this balance minimizes adverse effects like excessive or proarrhythmia. Natural sodium channel blockers, such as (TTX) produced by pufferfish, exemplify evolutionary adaptations for predation and defense, where TTX serves as a potent to deter predators by paralyzing excitable tissues, thereby informing modern through insights into high-affinity, selective inhibition. This evolutionary context highlights how toxin-derived mechanisms have been harnessed to develop synthetic blockers that mimic state-specific blockade for therapeutic precision.

Sodium Channel Biology and Blockade

Voltage-Gated Sodium Channel Structure

Voltage-gated sodium channels () are integral membrane proteins essential for the initiation and propagation of action potentials in excitable cells. These channels form a heterotetrameric complex consisting of a principal pore-forming α subunit and one or more auxiliary β subunits. The α subunit, encoded by genes in the SCN family, is a large polypeptide of approximately 2000 that folds into a pseudotetrameric with four homologous (). Each comprises six transmembrane segments (S1–S6), where S1–S4 form the voltage-sensing (VSD) and S5–S6 contribute to the (). The β subunits, encoded by SCN1B–SCN4B genes, are smaller glycoproteins with a single transmembrane segment and an extracellular immunoglobulin-like ; they modulate channel gating, trafficking, and . Key functional regions within the α subunit include the S4 segment of the VSD, which acts as the primary voltage sensor due to its positively charged arginine and lysine residues that respond to changes in membrane potential. The selectivity filter, located in the extracellular P-loops between S5 and S6 of each domain, confers Na⁺ specificity through the conserved DEKA motif (aspartate in DI, glutamate in DII, lysine in DIII, and alanine in DIV), which partially dehydrates and coordinates permeating Na⁺ ions while excluding other cations like K⁺. Fast inactivation is mediated by the intracellular linker between DIII and DIV, which contains the IFM (isoleucine-phenylalanine-methionine) motif that binds within the pore to occlude ion flow shortly after channel opening. Mammalian Nav channels exhibit isoform diversity with nine functional α subunit isoforms (Nav1.1–Nav1.9), each encoded by a distinct and displaying tissue-specific expression patterns that underlie specialized physiological roles. These isoforms share over 70% sequence identity but differ in gating properties, expression levels, and associations due to variations in their intracellular loops and C-termini. For instance, Nav1.5 predominates in cardiac myocytes, while Nav1.7, Nav1.8, and Nav1.9 are enriched in peripheral sensory neurons involved in signaling. Mutations in these genes are linked to a spectrum of channelopathies, including epilepsies, cardiac arrhythmias, and disorders.
IsoformGenePrimary Tissue ExpressionAssociated Diseases
Nav1.1SCN1ACNS (e.g., GABAergic interneurons)Epilepsy (e.g., GEFS+, Dravet syndrome)
Nav1.2SCN2ACNS (e.g., axons, dendrites)Epilepsy, autism spectrum disorder
Nav1.3SCN3ACNS (embryonic/neonatal)Neuropathic pain (potential role)
Nav1.4SCN4ASkeletal muscleMyotonia, periodic paralysis
Nav1.5SCN5ACardiac muscleArrhythmias (e.g., long QT syndrome, Brugada syndrome)
Nav1.6SCN8ACNS, PNS (e.g., nodes of Ranvier)Epilepsy, ataxia
Nav1.7SCN9APNS (sensory neurons, DRG)Pain disorders (e.g., erythromelalgia, congenital insensitivity to pain)
Nav1.8SCN10APNS (sensory neurons, DRG)Painful neuropathies, potential cardiac conduction defects
Nav1.9SCN11APNS (sensory neurons, DRG)Pain hypersensitivity, small fiber neuropathy
Nav channels cycle through distinct gating states—resting, open, and inactivated—to regulate Na⁺ influx during . In the resting state, at negative potentials, the gate (formed by the S6 segments) is closed, preventing ion permeation despite the inactivation gate being open. Upon , voltage-dependent movement of the S4 segments opens the gate, transitioning the channel to the open state for rapid Na⁺ conduction. Fast inactivation then occurs within milliseconds as the IFM motif plugs the intracellular mouth of the pore, rendering the channel nonconductive; recovery from inactivation requires to restore the resting state. These voltage- and time-dependent transitions ensure brief, self-limiting Na⁺ currents critical for shape.

Modes and Sites of Blockade

Sodium channel blockers exhibit distinct modes of with voltage-gated sodium channels, primarily categorized as , use-dependent, and state-dependent . refers to a constant, low-affinity inhibition that occurs independently of channel activity, resulting from baseline binding to channels in the resting state during low-frequency or single stimulations. In contrast, use-dependent involves enhanced inhibition during repetitive channel activations, where the drug preferentially accumulates in channels that undergo frequent opening and inactivation, due to slower dissociation rates relative to the stimulation interval. State-dependent encompasses variations in drug affinity across the channel's conformational states—resting (closed), open, and inactivated—with highest binding typically to the open or inactivated states, as described by the modulated receptor hypothesis. Binding kinetics govern these modes through association (k_on) and dissociation (k_off) rates, which determine the onset and recovery from blockade. For use-dependent block, the fraction of channels blocked can be approximated by the equation for drug association during exposure: fraction blocked = 1 - exp(-k_on \cdot [drug] \cdot t), where [drug] is the drug concentration and t is the time of exposure or effective pulse duration; recovery from block follows a time constant τ = 1/k_off, influencing the degree of accumulation over successive activations. These kinetics ensure that blockade intensifies with faster association to activated states and slower unbinding, amplifying inhibition under high-frequency conditions. Several factors modulate the extent of blockade. Membrane potential affects state transitions, with depolarization favoring inactivated states and thereby enhancing state-dependent binding, as channels spend more time in high-affinity conformations. Firing rate directly influences use-dependent block, as shorter intervals between action potentials limit recovery, leading to progressive channel occupancy. Additionally, pH alters blockade via protonation of the drug; acidic conditions promote the charged form of local anesthetic-type blockers, increasing their affinity for the inactivated state and potentiating use-dependent inhibition, while neutral forms at higher pH may favor tonic access through hydrophobic pathways. Some agents display reverse use-dependence, where blockade diminishes at higher frequencies due to rapid kinetics or state preferences that allow quicker recovery.06025-1) Pharmacologically, these modes enable selective suppression of abnormal electrical activity. Use- and state-dependent preferentially targets pathological high-frequency firing, such as in ischemic tissues where and exacerbate channel inactivation, thereby reducing excitability in hyperexcitable regions while sparing normal low-rate conduction. This selectivity underlies the therapeutic utility of sodium channel blockers in conditions involving aberrant repetitive activity.

Classification by Binding Site

Extracellular Blockers

Extracellular blockers of voltage-gated sodium channels primarily interact with site 1, located in the external near the selectivity filter at the pore entrance, where they achieve high-affinity, voltage-independent occlusion of the sodium permeation pathway. This binding physically plugs the extracellular mouth of the channel, preventing Na⁺ ions from entering without altering the channel's gating mechanism, and operates non-competitively with agents that access intracellular sites. The high affinity stems from electrostatic and hydrogen-bonding interactions between the toxin's positively charged groups and negatively charged residues in the channel's P-loops, effectively mimicking aspects of the Na⁺ hydration shell to occlude ion flow. Prominent examples include (TTX), derived from pufferfish and other marine organisms, and (STX), produced by dinoflagellates and accumulated in . TTX potently blocks neuronal isoforms such as Nav1.1, Nav1.2, Nav1.3, Nav1.4, Nav1.6, and Nav1.7 with dissociation constants (K_d) in the range of 1-10 nM, while STX exhibits similar nanomolar affinity (K_d ≈ 0.5-1 nM) for these TTX-sensitive channels. The molecular structure of TTX features multiple guanidinium groups that coordinate with the channel's selectivity filter residues, forming a tight seal that replicates the hydrated Na⁺ ion's interactions and halts conduction with near-irreversible kinetics under physiological conditions. STX shares this guanidinium-based but includes additional carbamoyl and hydroxyl groups that enhance its binding stability through hydrogen bonding in the outer vestibule. μ-Conotoxins, peptide toxins from venoms, represent another class of extracellular pore blockers, exemplified by μ-conotoxins GIIIA, KIIIA, and PIIIA, which insert into site 1 via their cationic residues and cysteine-stabilized framework. These peptides electrostatically impede Na⁺ passage by occluding the entrance, often with isoform selectivity; for instance, KIIIA inhibits Nav1.7 with an IC₅₀ in the low nanomolar range, differing from the broader spectrum of small-molecule guanidinium toxins. Like TTX and STX, μ-conotoxins exhibit slow dissociation rates, contributing to their prolonged blockade. Due to their potent toxicity and narrow therapeutic windows, extracellular blockers like TTX, STX, and μ-conotoxins are primarily employed as research tools to probe sodium channel function, isoform specificity, and permeation dynamics rather than in clinical settings.

Intracellular Blockers

Intracellular sodium channel blockers access their binding sites from the cytoplasmic side of the , primarily entering through lateral fenestrations—hydrophobic portals in the lipid-facing regions of the —and interacting within the central inner cavity. These agents engage the local anesthetic receptor sites, particularly sites 2 through 4, via hydrophobic interactions with residues lining the S6 transmembrane segments, especially in domains III and IV. Prominent examples of such blockers include quaternary amines like QX-314, a permanently charged lidocaine derivative that is membrane-impermeant and thus functions as a selective for probing intracellular mechanisms, requiring co-application with permeabilizing agents to enter cells and bind sodium channels. In contrast, tertiary amines such as lidocaine gain intracellular access in their protonated form primarily through the open channel pore, enabling blockade from the cytoplasmic vestibule. The mechanism of these blockers involves preferential binding to inactivated channel states, with access modulated by voltage and pH; the unionized (neutral) form crosses the more readily, while the protonated (ionized) species delivers the potent electrostatic and hydrophobic interactions at the receptor site. Recovery from blockade is relatively rapid, with time constants typically ranging from 100 to 500 ms for agents like lidocaine, allowing during . Intracellular blockers demonstrate general state-dependence, favoring open and inactivated conformations over resting states. These agents exhibit unique properties such as frequency-dependent accumulation, where repeated depolarizations enhance in hyperexcitable pathological conditions like ischemia or , and allosteric of gating that stabilizes the inactivated state and alters voltage sensor movements.

Blockers with Unclear Mechanisms

Certain sodium channel blockers exhibit inhibitory effects on voltage-gated sodium channels () without a fully elucidated or precise , complicating their relative to agents with well-defined extracellular or intracellular interactions. These compounds often demonstrate partial or low-affinity , potentially involving allosteric or secondary effects that do not align with traditional pore-blocking profiles. Research into these agents highlights ongoing uncertainties in , particularly as structural studies reveal isoform-specific variations. Riluzole, primarily used in (ALS), partially inhibits Nav channels by blocking persistent sodium currents (I_NaP) in a dose-dependent manner, with an of approximately 2 μM observed in mammalian neurons. Patch-clamp studies indicate that riluzole preferentially targets the inactivated state of TTX-sensitive sodium channels at depolarized potentials, but its exact interaction site remained ambiguous until recent structural analyses. A 2024 high-resolution and NMR study revealed riluzole binding within intramembrane fenestrations of prokaryotic NavMs channels, stabilizing the inactivated state through an allosteric mechanism rather than direct pore occlusion, though this finding requires further validation in eukaryotic isoforms. Proposed mechanisms include allosteric shifts in channel gating or off-target interactions, as evidenced by incomplete in heterogeneous isoform responses and variable potency across neuronal types. Zonisamide, an antiepileptic drug, exerts low-affinity inhibition on voltage-sensitive sodium channels, reducing sustained high-frequency repetitive firing without fully conforming to classic use-dependent blockade patterns. Electrophysiological data suggest it alters the fast inactivation threshold of channels, but the precise and contribution to overall antiseizure activity remain unclear, with mixed effects potentially involving concurrent modulation. Patch-clamp recordings demonstrate partial suppression of sodium currents in neuronal models, supporting hypotheses of allosteric or indirect interactions rather than high-affinity site-specific binding. Key research challenges for these blockers stem from Nav isoform heterogeneity, where riluzole and zonisamide show differential efficacy across Nav1.x subtypes, complicating mechanistic attribution in complex tissues. Recent 2025 computational and enhanced sampling studies on Nav1.5 channels indicate diverse access routes and binding poses for various inhibitors, suggesting novel or lipid-facing sites, yet these remain inconclusive for multifunctional agents like riluzole and zonisamide due to limited isoform-specific structural data. Unlike site-specific blockers, these compounds often display lower potency (e.g., IC50 values in the micromolar range) and multifunctional profiles; for instance, riluzole additionally inhibits glutamate release from presynaptic terminals, contributing to its neuroprotective effects beyond Nav modulation.

Therapeutic Uses in Cardiology

Class Ia Agents

Class Ia agents are a subclass of antiarrhythmic drugs characterized by moderate blockade of voltage-gated s, coupled with effects on channels that prolong the () and on the electrocardiogram. These agents exhibit intermediate kinetics, with moderate on- and off-rates of binding, leading to use-dependent blockade that is more pronounced at higher heart rates. This profile distinguishes them from other Class I agents by balancing conduction slowing with prolongation, though their use has declined due to proarrhythmic risks. The prototypical Class Ia agent, quinidine, was first employed clinically in 1914 for arrhythmia suppression, marking the advent of modern antiarrhythmic therapy. Other key drugs include and . Quinidine is typically administered orally at 200-400 mg every 6 hours or intravenously at similar doses for acute settings, with primary elimination via hepatic through (about 80%) and partial renal clearance (20%). dosing ranges from 3-6 g daily orally or 10-17 mg/kg intravenously over 30-60 minutes, undergoing hepatic to its N-acetylprocainamide (NAPA), with significant renal clearance for both parent drug and metabolite, necessitating dose adjustments in renal impairment. is given at 100-200 mg every 6 hours orally, featuring properties and predominantly renal elimination (50-80%), with half-life prolongation in renal dysfunction. All three agents require to avoid , given their narrow therapeutic indices. In the heart, Class Ia agents slow conduction velocity in atrial and ventricular myocardium by inhibiting phase 0 sodium influx, particularly during rapid rhythms due to their use-dependent binding, which preferentially affects ischemic or depolarized tissue. This mechanism terminates reentrant arrhythmias by prolonging the and is effective against both supraventricular tachycardias (SVT) and ventricular tachycardias (VT). However, their blockade extends , increasing the risk of . Specific indications for Class Ia agents center on (AF), including pharmacological and maintenance of in patients without significant structural heart disease (SHD). Intravenous is recommended (Class 1 recommendation) for hemodynamically stable preexcited AF and may be considered (Class 2b) as an alternative for when other agents are unsuitable, particularly in with normal hearts. Quinidine and support rhythm control in select non-SHD cases, though evidence is historical. The 2023 ACC/AHA/ACCP/HRS guidelines caution against their use in AF with SHD (e.g., , , LVEF ≤40%), due to heightened proarrhythmic and mortality risks, favoring or safer alternatives like . No major updates endorsed expanded Class Ia roles in 2023, emphasizing early rhythm control strategies.

Class Ib Agents

Class Ib agents are a subclass of antiarrhythmic drugs within the Vaughan-Williams classification that exhibit weak blockade of voltage-gated sodium channels, characterized by fast association and dissociation kinetics with a recovery (tau) typically less than 1 second. This rapid on/off binding results in minimal effects on duration (APD) and QT interval in normal myocardium, with a slight shortening of APD in ventricular tissue, and demonstrates reverse use-dependence, whereby their effects diminish at higher heart rates. These properties make them particularly suitable for targeting arrhythmias in compromised cardiac tissue without significantly impairing conduction in healthy cells. Key representatives include lidocaine, administered intravenously for acute management of ventricular arrhythmias; , an oral analog of lidocaine used for chronic suppression; and , which shares similar blocking actions. Lidocaine is typically given as an initial bolus of 1-1.5 mg/kg over 2-3 minutes, followed by additional doses of 0.5-0.75 mg/kg if needed, up to a total of 3 mg/kg, with an elimination of approximately 1.5-2 hours. has a longer of 10-12 hours, allowing for twice- or thrice-daily oral dosing, while 's in antiarrhythmic use ranges from 7-42 hours, often requiring dose adjustments for hepatic . In the heart, Class Ib agents preferentially bind to and block sodium channels in ischemic or depolarized myocardium, where elevated extracellular levels during ischemia enhance drug affinity and promote inactivation of channels in a use-dependent manner. This selective action suppresses ectopic activity in post-infarction and ventricular cells, slightly shortening ventricular without prolonging refractoriness in normal tissue. These agents are primarily indicated for the acute treatment of ventricular tachycardia (VT) and ventricular fibrillation (VF) following myocardial infarction (MI), where they effectively suppress life-threatening arrhythmias originating from ischemic zones. Clinical evidence supports their role in reducing the incidence of recurrent ventricular arrhythmias in the early post-MI period, with studies demonstrating a decrease in sudden cardiac death risk when used prophylactically or therapeutically in high-risk patients. As of 2025, updated guidelines continue to endorse intravenous lidocaine as a second-line option for hemodynamically stable VT in the post-MI setting, based on ongoing reviews of trial data emphasizing its safety profile in ischemic contexts.

Class Ic Agents

Class Ic agents represent the subgroup of sodium channel blockers with the most potent inhibition of cardiac sodium currents, exhibiting very slow unbinding kinetics from the sodium channel with recovery time constants exceeding 10 seconds, such as approximately 19 seconds for flecainide. These drugs demonstrate marked use-dependence, whereby blockade intensifies with faster heart rates due to preferential binding during channel activation or inactivation states, enhancing their efficacy during tachyarrhythmias. Unlike other subclasses, Class Ic agents lack significant effects on potassium channels, resulting in no prolongation of the QT interval or action potential duration. The primary Class Ic agents are and , both used for rhythm control in supraventricular arrhythmias. is typically initiated at 50 mg twice daily (BID), titrated up to 150 mg BID based on response and tolerability, while propafenone starts at 150 mg three times daily (TID), increasing to 300 mg TID if needed. 's metabolism is predominantly mediated by the enzyme , leading to variable influenced by genetic polymorphisms in this enzyme, which can result in higher plasma levels and potential toxicity in poor metabolizers. In cardiac tissue, Class Ic agents exert their antiarrhythmic effects by profoundly depressing phase 0 depolarization, thereby slowing conduction velocity across atrial and ventricular myocardium without substantially altering refractoriness or duration. This selective prolongation of conduction time disrupts reentrant circuits, particularly in atrial tissue, making them highly effective for terminating and preventing paroxysmal supraventricular tachycardias. Per the 2024 () guidelines, and are recommended as first-line options for pharmacological and long-term rhythm control in patients with paroxysmal (AF) or who have no underlying structural heart disease, such as preserved left ventricular (>40%) and absence of or significant valvular abnormalities. However, the Cardiac Suppression Trial () conducted in 1989 demonstrated that (and encainide) increased mortality risk in post-myocardial infarction patients with asymptomatic ventricular arrhythmias, leading to contraindications for their use in ischemic or structurally compromised hearts.

Other Therapeutic Applications

Local Anesthetics

Local anesthetics represent a key subclass of sodium channel blockers employed in regional anesthesia to interrupt nerve conduction and provide targeted pain relief. These agents are chemically classified into two primary groups based on their structure: amino-amides, such as lidocaine and bupivacaine, which undergo hepatic metabolism, and amino-esters, like procaine, which are hydrolyzed by plasma esterases. Both types function as weak bases with pKa values ranging from approximately 7.7 to 9.0, enabling pH-dependent activity where the un-ionized form facilitates membrane penetration into nerve axons, and the subsequently ionized form binds intracellularly to voltage-gated sodium channels, preventing sodium influx and action potential propagation. In peripheral nerves, local anesthetics exert a differential blockade, selectively inhibiting smaller-diameter fibers before larger ones due to variations in fiber geometry, myelination, and density. Non-myelinated C-fibers, which transmit signals, are particularly sensitive and blocked first, followed by autonomic and sensory fibers, with motor fibers affected last. This hierarchical effect supports sensory analgesia with relative sparing of motor function at lower concentrations. Blockade intensity is concentration-dependent; for instance, 1-2% lidocaine solutions are standard for infiltration , achieving rapid onset within minutes. Clinically, these agents are administered via spinal or epidural routes for intraoperative and postoperative analgesia, or through peripheral blocks for procedures on . Lidocaine provides short-duration lasting 1-2 hours, ideal for brief interventions, whereas bupivacaine offers prolonged effects of 4-8 hours, suitable for major surgeries. By 2025, ultrasound-guided techniques have revolutionized these applications, enabling real-time visualization for precise perineural injection, which reduces required doses by up to 50% and lowers complication risks compared to landmark-based methods. Bupivacaine poses a notable risk, stemming from its high-affinity, slow-dissociation of the cardiac isoform Nav1.5, which can precipitate arrhythmias during inadvertent systemic . To mitigate rapid uptake and extend block duration, epinephrine is commonly added as a vasoconstrictor at concentrations of 1:200,000 to 1:100,000, inducing local ischemia that slows anesthetic diffusion into circulation.

Anticonvulsants and Antiepileptics

Sodium channel blockers play a central role in the management of , particularly for focal seizures, by modulating neuronal excitability through inhibition of voltage-gated sodium channels (VGSCs). These agents, including , , and , preferentially target neuronal isoforms such as Nav1.1 and Nav1.2 to suppress hyperexcitability in epileptogenic foci. By stabilizing neuronal membranes and reducing the initiation or propagation of action potentials, they effectively dampen abnormal synchronous firing characteristic of seizures. Carbamazepine exerts a use-dependent of VGSCs, particularly on Nav1.1 and Nav1.2, inhibiting repetitive neuronal firing during sustained depolarization while sparing normal synaptic transmission. similarly blocks sodium channels in a voltage- and use-dependent manner, with relative selectivity for neuronal isoforms that spares significant inhibition of the cardiac Nav1.5 channel, minimizing cardiovascular risks. In contrast, uniquely enhances slow inactivation of VGSCs without substantially affecting fast inactivation, selectively reducing channel availability during prolonged high-frequency activity. Clinically, these drugs are indicated for focal seizures and, in the case of , associated with in patients. is typically initiated at low doses and titrated to a range of 200-1200 mg/day in divided doses, with autoinduction of its metabolism via necessitating dose adjustments after 2-4 weeks to achieve steady-state levels. and are often used as adjunctive therapies; for instance, the once-daily extended-release formulation of (Motpoly XR) received FDA approval in 2024 for adjunctive use in primary generalized tonic-clonic seizures in adults and aged 4 years and older. Randomized controlled trials (RCTs) demonstrate that sodium channel blockers achieve reductions of 50-70% in responsive patients with focal , with responder rates (≥50% reduction) around 55% for add-on therapies like zonisamide and similar for in monotherapy. However, a serious of is the risk of Stevens-Johnson syndrome (SJS), particularly in Asian populations carrying the HLA-B*1502 allele, where screening is recommended prior to initiation to prevent severe cutaneous reactions.

Analgesics and Pain Management

Sodium channel blockers play a significant role in managing neuropathic and inflammatory by targeting voltage-gated sodium channels in peripheral sensory neurons, particularly those involved in aberrant signaling following . These agents primarily act by inhibiting ectopic firing in damaged nerves, where upregulated sodium channels contribute to spontaneous activity and heightened excitability. Specifically, isoforms such as Nav1.7, Nav1.8, and Nav1.9, which are predominantly expressed in (DRG) neurons, are critical for transduction and propagation; blocking these channels disrupts the initiation and transmission of nociceptive signals. Additionally, sodium channel blockade can reduce central sensitization phenomena, such as wind-up in the spinal dorsal horn, where repeated nociceptive inputs amplify neuronal responses. Their use-dependent blockade preferentially affects hyperexcitable states in injured nerves, enhancing selectivity for pathological over normal signaling. Among established sodium channel blockers used in pain management, mexiletine is employed off-label for chronic neuropathic conditions, including diabetic peripheral neuropathy and postherpetic neuralgia, due to its oral bioavailability and ability to suppress peripheral nerve hyperexcitability. Typical dosing for mexiletine in these indications ranges from 150 to 300 mg three times daily, titrated based on tolerance and efficacy, with monitoring for gastrointestinal and cardiovascular side effects. Eslicarbazepine acetate, primarily approved for epilepsy, has been investigated for neuropathic pain, including trigeminal and peripheral diabetic neuropathy, by modulating sodium channel activity to alleviate allodynia and hyperalgesia, though evidence remains limited to small studies and open-label data, with mixed results in larger trials. Clinical evidence supports the utility of these blockers in specific syndromes. For instance, has been associated with significant pain relief in , with studies reporting reductions in pain intensity and improved in responsive patients. In , both and investigational blockers have shown promise in reducing spontaneous pain and evoked . A 2021 of pharmacotherapeutic options for , including modulators, indicated responder rates of 30-50% for substantial pain reduction (≥30% from baseline), underscoring their role in . These agents exhibit opioid-sparing potential by providing independent , thereby reducing reliance on opioids in management and mitigating risks of dependence and overdose. Guidelines for non-opioid pain management emphasize sodium channel blockers as viable options for chronic , particularly in refractory cases, aligning with broader efforts to prioritize non-addictive alternatives amid the opioid crisis.

Adverse Effects and Toxicity

Cardiovascular and Neurological Effects

Sodium channel blockers exert significant adverse effects on the cardiovascular system, primarily through their inhibition of cardiac sodium influx, which slows conduction and can precipitate proarrhythmia. Class Ia agents, such as quinidine and , are particularly associated with due to their prolongation of the action potential duration and , increasing the risk of polymorphic . and are common manifestations across classes, resulting from depressed and , potentially leading to in severe cases. For class Ic agents like , toxicity is indicated by QRS complex widening exceeding 100 ms on electrocardiogram, reflecting profound conduction delay and heightened risk of ventricular arrhythmias. These state-dependent blocking effects are amplified during rapid heart rates or ischemic conditions, exacerbating proarrhythmic potential. Neurological effects of sodium channel blockers arise from their blockade of neuronal sodium channels, leading to , particularly at high doses. Symptoms include , , and , progressing to seizures and in overdose scenarios, as the drugs suppress neuronal excitability and repetitive firing. This toxicity is pH-dependent, with worsening the sodium channel blockade by increasing the drugs' binding affinity in their charged state, thereby intensifying both cardiac and neurological manifestations. Risk factors for these adverse effects include drug interactions that elevate sodium channel blocker levels, such as co-administration with CYP3A4 or P-glycoprotein inhibitors, which impair metabolism and clearance. Comorbidities like heart failure further heighten vulnerability by altering drug pharmacokinetics and exacerbating conduction abnormalities. Recent data indicate a proarrhythmic risk of approximately 4-5% with flecainide use in atrial fibrillation management, underscoring the need for cautious application in susceptible patients. Monitoring involves serial electrocardiograms to detect prolongation, QRS widening, and extension, which signal emerging toxicity and guide dose adjustments. For specifically, therapeutic drug monitoring of serum levels (target procainamide 4–10 μg/mL and combined procainamide plus N-acetylprocainamide 10–30 μg/mL) is essential to prevent accumulation and adverse events.

Toxicity Management

Management of sodium channel blocker toxicity begins with recognizing the clinical presentation, which typically includes widened on electrocardiogram (>100 ms), (VT) or (VF), and seizures, collectively manifesting as a sodium channel blocker toxicity characterized by wide-complex tachycardia refractory to standard advanced cardiovascular (ACLS) protocols. This arises from profound blockade leading to impaired cardiac conduction and excitability, often compounded by and . Primary treatments target reversal of sodium channel blockade and hemodynamic stabilization. is administered intravenously at 1-2 mEq/kg to address and provide a sodium load that enhances channel recovery, particularly effective in cases with QRS prolongation or arrhythmias; a continuous (1.5-2 times maintenance rate) follows to maintain pH of 7.45-7.55. For cardiotoxic agents such as bupivacaine, emulsion therapy involves a 20% intralipid bolus of 1.5 mL/kg over 1 minute, followed by an of 0.25 mL/kg/min until stability, leveraging partitioning to reduce free drug levels. exacerbates sodium channel binding, and alkalinization with mitigates this effect. Supportive measures are essential and include immediate with endotracheal if respiratory depression or altered mental status occurs, alongside benzodiazepines for control to avoid exacerbating cardiac instability. may be considered for renally cleared agents like lidocaine, though extraction efficiency is low due to high protein binding and , limiting its overall utility. For refractory cardiovascular collapse, (ECMO) provides circulatory support and may be reasonable in cases unresponsive to conventional therapies, as per the 2025 guidelines. Hypertonic saline (e.g., 3% at 2-5 mL/kg) may be considered in refractory cases after optimizing bicarbonate and fluids, for additional sodium loading. Evidence from case series indicates survival rates of approximately 77% for class I antiarrhythmic toxicity with early intervention combining these strategies, particularly when initiated prior to cardiac arrest.

Research and Future Directions

Selective Isoform-Targeted Blockers

Ongoing research into selective isoform-targeted blockers for voltage-gated sodium channels (Nav) aims to enhance therapeutic precision by minimizing off-target effects on non-target isoforms, thereby improving safety profiles in conditions like pain and cardiac arrhythmias. These efforts focus on exploiting structural differences among the nine Nav isoforms, particularly in their voltage-sensing domains and pore regions, to develop compounds with high specificity. Key target isoforms include Nav1.7, which is predominantly expressed in peripheral nociceptors and implicated in inherited pain disorders such as ; Nav1.8, associated with inflammatory and signaling in neurons; and Nav1.5, the primary cardiac isoform responsible for initiation, where selectivity helps avoid neuronal side effects. Significant advancements include small-molecule inhibitors and biologics designed for isoform specificity. For Nav1.7, clinical candidates like ANP-230, a state-independent blocker also targeting Nav1.8 and Nav1.9, have entered testing for broad-spectrum analgesia, demonstrating superior efficacy over in preclinical models. Biologics such as monoclonal antibodies against Nav1.7 have shown promise in blocking signals without penetration, reducing risks of . For Nav1.8, suzetrigine (VX-548) exemplifies high selectivity, with over 30,000-fold preference over other Nav isoforms, and received FDA approval in January 2025 for moderate-to-severe acute in adults following positive phase 3 results in reducing intensity after and bunionectomy surgeries. In the cardiac domain, novel derivatives selectively inhibit Nav1.5 in atrial cardiomyocytes, prolonging the to suppress arrhythmias without affecting ventricular conduction. Additionally, CRISPR-based genetic screens have identified potential allosteric modulators by systematically evaluating variants in Nav1.2 and related isoforms, revealing hotspots for subtype-specific binding that inform . The primary advantages of these selective blockers lie in their potential to mitigate adverse effects common to non-selective agents, such as cardiac conduction abnormalities from neuronal-targeted therapies. For instance, Nav1.7 and Nav1.8 inhibitors avoid blocking Nav1.5, preserving normal heart rhythm while alleviating . Structure-based has been accelerated by high-resolution cryo-EM structures of Nav1.7, achieving resolutions below 3 Å, which elucidate binding pockets for allosteric sites and enable rational optimization of selectivity. Preclinical studies report 10- to 100-fold selectivity ratios for , correlating with reduced potassium channel interference and improved therapeutic windows. Despite these progresses, challenges persist due to the high among Nav isoforms, approximately 80% in the transmembrane domains, which complicates achieving exquisite selectivity and often requires targeting extracellular or allosteric sites. Blood-brain barrier penetration remains a hurdle for central indications, as many selective small molecules exhibit poor CNS , limiting their utility in neuropathic conditions. Furthermore, while preclinical data demonstrate 10-fold or greater selectivity in systems, translation to human efficacy has been inconsistent, with some Nav1.7 inhibitors failing phase 2 trials due to insufficient analgesia despite potent blockade.

Emerging Therapeutic Applications

Recent research has explored the potential of analogs as neuroprotective agents in by modulating activity to mitigate and neuronal damage. For instance, a novel riluzole-based compound, VA945, demonstrates enhanced inhibition of voltage-dependent sodium channels, offering improved neuroprotective effects compared to in preclinical models of ischemic injury. Similarly, partial blockers targeting Nav1.7 channels are under investigation for prophylaxis, aiming to reduce hyperexcitability that contributes to migraine attacks. Preclinical studies indicate that Nav1.7 inhibition in trigeminal ganglia cultures decreases neuronal firing rates, suggesting a role in preventing migraine initiation. In , particularly associated with , Nav1.6 channels have emerged as a key target due to their role in enhancing tumor-induced neuronal hyperexcitability and signaling. Exosomes from glioma cells upregulate Nav1.6 expression in sensory neurons via TNF-α, leading to increased sensitivity; blocking Nav1.6 has shown promise in reducing this hyperexcitability in rodent models. Furthermore, Nav1.6 promotes glioma progression by regulating ion and activating oncogenic pathways like AKT/ERK, highlighting its dual role in tumor growth and associated . Clinical trials have advanced Nav1.8 inhibitors for conditions. VX-150, a selective Nav1.8 blocker, demonstrated significant relief in phase 2 studies for of the . Suzetrigine (VX-548), approved in 2025 for acute , showed clinically meaningful reductions in scores in phase 3 trials for postoperative , with ongoing evaluations for potential expansion to other indications. In (ALS), extensions of therapy combined with modulation are being tested to enhance ; real-world data from 2025 analyses show slows functional decline, and adjunctive blockers may amplify this by reducing excitotoxic sodium influx in motor neurons. Translational advances include for sodium channel blockers, with 2024-2025 publications detailing platforms that accelerate identification of isoform-selective inhibitors for . These tools, such as RosettaVS, predict affinities with high accuracy, enabling rapid optimization of candidates targeting Nav1.7 and Nav1.8. Combination therapies pairing sodium channel blockers with or biologics are also gaining traction, reducing opioid requirements while enhancing analgesia; for example, Nav1.8 inhibitors like suzetrigine provide additive effects in postoperative models without increasing adverse events. Prospects for sodium channel blockers include favorable regulatory outlooks, with the FDA granting fast-track designations in 2025 for agents like LTG-001 in acute and rare disorders, expediting development for unmet needs in neuropathic conditions. Ethical considerations in use emphasize balancing long-term efficacy against risks like cardiac conduction delays, necessitating and monitoring to ensure in extended therapies.

References

  1. [1]
    Physiology, Sodium Channels - StatPearls - NCBI Bookshelf
    The prevention of nerve conduction is the mechanism of drugs like lidocaine blocks the potentiation of pain signals, while antiepileptics such as phenytoin and ...
  2. [2]
    Mechanism of sodium channel block by local anesthetics ... - NIH
    Local anesthetics, antiarrhythmics, and anticonvulsants include both charged and electroneutral compounds that block voltage-gated sodium channels.
  3. [3]
    Sodium Channel Blocker Toxicity - StatPearls - NCBI Bookshelf
    Mar 2, 2024 · Sodium channel blockers cross the blood-brain barrier and function by multiple mechanisms. They inhibit the gamma-aminobutyric acid (GABA) ...
  4. [4]
    Sodium Channel Blocker - an overview | ScienceDirect Topics
    A sodium channel blocker is defined as a substance that inhibits voltage-gated sodium channels on cell membranes, thereby affecting their activity, which is ...
  5. [5]
    The Sodium Channel as a Target for Local Anesthetic Drugs - Frontiers
    Oct 31, 2011 · Na channels are the source of excitatory currents for the nervous system and muscle. They are the target for a class of drugs called local anesthetics (LA).<|separator|>
  6. [6]
    Sodium Channels and Local Anesthetics—Old Friends With New ...
    The long history of local anesthetics (LAs) starts out in the late 19th century when the content of coca plant leaves was discovered to alleviate pain.
  7. [7]
    Sodium Channel Blocker - an overview | ScienceDirect Topics
    Sodium channel blockers work by inhibiting sodium influx via cell membrane, thus dampening the rate of rapid depolarization, and reducing cell excitability.
  8. [8]
    Neurological perspectives on voltage-gated sodium channels - PMC
    The activity of voltage-gated sodium channels has long been linked to disorders of neuronal excitability such as epilepsy and chronic pain.
  9. [9]
    Recent Developments Regarding Voltage-Gated Sodium ... - Frontiers
    Recent developments regarding voltage-gated sodium channel blockers for the treatment of inherited and acquired neuropathic pain syndromes.
  10. [10]
    Global surge in serious heart rhythm disorders sparks urgent call to ...
    Mar 1, 2025 · 1 in 3 people worldwide will develop a potentially life-threatening heart rhythm disorder in their lifetime, yet awareness remains critically low.
  11. [11]
    Therapeutic efficacy of voltage-gated sodium channel inhibitors in ...
    Jun 28, 2023 · ASMs acting as sodium channel inhibitors stabilize sodium channels by preventing them to return to the active state and potentiating the ...
  12. [12]
    Sodium channel blockers for the treatment of neuropathic pain - PMC
    Drugs that block voltage-gated sodium channels are efficacious in the management of neuropathic pain.Missing: definition | Show results with:definition
  13. [13]
    Druggability of Voltage-Gated Sodium Channels—Exploring Old ...
    The state-dependent block allows local anesthetics (LAs) and anti-arrhythmic drugs (AADs) to selectively prevent generations of high-frequency action potentials ...
  14. [14]
    The mechanism of non‐blocking inhibition of sodium channels ...
    Jan 15, 2021 · The trick that enables them to do so is state-dependence, that is, their preference for certain conformational states of the channel protein.
  15. [15]
    An overview of drug‐induced sodium channel blockade and ...
    Dec 11, 2024 · These drugs are often used in the acute management of ventricular arrhythmias. In contrast, Class Ia agents, such as quinidine, block Na+ ...
  16. [16]
    From Poison to Promise: The Evolution of Tetrodotoxin and Its ...
    Jul 24, 2021 · ... Evolutionary Diversification of TTX-Resistant Sodium Channels in a Predator–Prey Interaction. Nature. 2005;434:759–763. doi: 10.1038 ...
  17. [17]
    The Chemical and Evolutionary Ecology of Tetrodotoxin (TTX ... - MDPI
    Skin secretions from Taricha [25,60,63,100–102], and Notophthalmus [103–109], are known to be lethal to (or deter predation by) potential predators. Reports ...
  18. [18]
    Voltage-Gated Sodium Channels: Structure, Function ...
    The tremendous therapeutic potential of voltage-gated sodium channels (Navs) has been the subject of many studies in the past and is of intense interest ...
  19. [19]
    Na+ Channel β Subunits: Overachievers of the Ion ... - Frontiers
    Sep 27, 2011 · Voltage gated Na+ channels (VGSCs) in mammals contain a pore-forming α subunit and one or more β subunits. There are five mammalian β ...Molecular Diversity and... · Regulation of Excitability by... · Alterations in VGSC...
  20. [20]
    Voltage-gated Na Channel Selectivity: The Role of the Conserved ...
    Cloning of the family of mammalian Na+ channel genes has identified four key residues of the selectivity region as an aspartate-glutamate-lysine-alanine (DEKA) ...
  21. [21]
    Overview of the voltage-gated sodium channel family
    Feb 24, 2003 · The voltage-gated sodium channel is a large, multimeric complex, composed of an α subunit and one or more smaller β subunits [3].
  22. [22]
    Inherited disorders of voltage-gated sodium channels - JCI
    Aug 1, 2005 · A variety of inherited human disorders affecting skeletal muscle contraction, heart rhythm, and nervous system function have been traced to mutations in genes ...Abstract · Introduction · Cardiac sodium channelopathies · Neuronal sodium...
  23. [23]
    Structures of closed and open states of a voltage-gated sodium ...
    Mar 27, 2017 · These two structures allow completion of a closed–open–inactivated conformational cycle in a single voltage-gated sodium channel and give ...
  24. [24]
    State-Dependent Inhibition of Sodium Channels by Local Anesthetics
    Local anesthetics inhibit sodium channels, with binding depending on channel conformation and state transitions, and the open and inactivated states have ...
  25. [25]
    Two components of use-dependent block of Na+ current by ...
    Na+ channel. Transition to drug-bound (blocked) or drug-free (unblocked) states can be expressed bythe rate constants of association and dissociation, which.
  26. [26]
    Protonation underlies tonic vs. use-dependent block - PNAS
    Mar 21, 2018 · The drug crosses the open activation gate in use-dependent inhibition, while it follows an alternative hydrophobic pathway in tonic block.
  27. [27]
    Sodium Channels and Local Anesthetics—Old Friends With New ...
    The long history of local anesthetics (LAs) starts out in the late 19th century when the content of coca plant leaves was discovered to alleviate pain.
  28. [28]
    Differential binding of tetrodotoxin and its derivatives to voltage ... - NIH
    Site 1 is located at the entrance of the pore overlapping the ion selectivity filter. Here, tetrodotoxin (TTX, 1, Figure 1), saxitoxin (STX) and μ‐conotoxin ...Missing: mu- | Show results with:mu-
  29. [29]
    Neurotoxins and Their Binding Areas on Voltage-Gated ... - Frontiers
    μ-Conotoxins such as KIIIA and GIIIA are strongly cationic peptides. Upon binding at site 1, these peptides electrostatically impede the outward movement of the ...Missing: mu- | Show results with:mu-
  30. [30]
    Structural basis for the modulation of voltage-gated sodium ...
    STX is a related guanidinium neurotoxin, produced by marine dinoflagellates and cyanobacteria, that competes with TTX for binding to Nav channels (15). The term ...Abstract · Recognition Of Ttx · Materials And Methods
  31. [31]
    Guanidinium Toxins and Their Interactions with Voltage-Gated ...
    These guanidinium groups confer high biological activity with high affinity and ion flux blockage capacity for voltage-gated sodium channels (NaV). Members of ...Missing: mimic | Show results with:mimic
  32. [32]
    μ-Conotoxins that differentially block sodium channels NaV1 ... - PNAS
    Jun 7, 2011 · μ-Conotoxins that differentially block sodium channels NaV1.1 through 1.8 identify those responsible for action potentials in sciatic nerve.
  33. [33]
    Energetic Localization of Saxitoxin in Its Channel Binding Site
    Saxitoxin (STX) selectively blocks the voltage-gated sodium channel at the outer vestibule lined by P-loops of the four domains.
  34. [34]
    Novel Interactions Identified between μ-Conotoxin and the Na + ...
    μ-Conotoxins (μ-CTX) are peptides that inhibit Na+ flux by blocking the Na+ channel pore. Toxin residue arginine 13 is critical for both high affinity ...
  35. [35]
    Voltage-Gated Sodium Channel Inhibition by µ-Conotoxins - MDPI
    µ-Conotoxins, characterised by a conserved cysteine-framework (CC–C–C–CC), potently inhibit NaV channel currents by blocking the pore of the NaV channel, ...
  36. [36]
    Fenestrations control resting-state block of a voltage-gated sodium ...
    Dec 5, 2018 · We show here that potency of local anesthetics (LAs) and antiarrhythmic drugs (AADs) that block sodium channels is controlled by fenestrations.
  37. [37]
    Structural Basis for Pharmacology of Voltage-Gated Sodium and ...
    These drugs bind to a common receptor site in the pore and impede ion permeation. The amino acid residues that form the receptor sites for sodium channel ...
  38. [38]
    Structural basis for antiarrhythmic drug interactions with the human ...
    Feb 6, 2019 · The results revealed that both antiarrhythmic and local anesthetic drugs share a receptor site formed by the S6 segments from DIII and DIV.
  39. [39]
    Co-application of Lidocaine and the Permanently Charged Sodium ...
    Nociceptive-selective local anesthesia is produced by entry of the permanently charged lidocaine-derivative QX-314 into nociceptors when coadministered with ...
  40. [40]
    Sidedness of Carbamazepine Accessibility to Voltage-Gated ... - NIH
    Similarly, studies examining pH dependence of action of lidocaine concluded that the protonated (cationic) form of the drug cannot directly access the sodium ...
  41. [41]
    Local anesthetics as effectors of allosteric gating. Lidocaine effects ...
    Our results support a reinterpretation of local anesthetic action whereby lidocaine functions as an allosteric effector to enhance Na channel inactivation. Full ...
  42. [42]
    Antiarrhythmic Medications - StatPearls - NCBI Bookshelf - NIH
    Indications · Class Ia: Causes moderate degree blockage of fast sodium channels. Drugs include quinidine, procainamide, and disopyramide. · Class Ib: Causes mild ...
  43. [43]
    Class I Antiarrhythmics (Sodium-Channel Blockers)
    Class IA and IC drugs bind to (and therefore block) sodium channels in both activated and inactivated states, which makes them particularly effective in ...Missing: stabilize | Show results with:stabilize
  44. [44]
    Modernized Classification of Cardiac Antiarrhythmic Drugs
    Oct 22, 2018 · Quinidine is additionally proarrhythmic under conditions of prolonged AP recovery, at least partially reflecting its additional IK-blocking ...Abstract · Modernized Scheme Based On... · Recapitulation
  45. [45]
    Blunderbuss to Mickey Mouse: The Evolution of Antiarrhythmic Targets
    Quinidine terminates episodes of palpitations, said a patient to Wenckebach in 1914 ... The decades after the first therapeutic application of quinidine saw the ...
  46. [46]
    Antiarrhythmics - EMCrit Project
    Jan 8, 2025 · Procainamide also has ganglionic blockade properties, which may cause hypotension when administered in high doses. Quinidine also has. Vagolytic ...
  47. [47]
    Therapeutic drug monitoring: antiarrhythmic drugs - PubMed Central
    Procainamide is usually given orally at a total dose of 3–6 g day−1. Bioavailability is high and peak plasma concentrations are achieved 1–2 h after tablet ...
  48. [48]
    Antiarrhythmics: elimination and dosage considerations in hepatic ...
    For sotalol, disopyramide and procainamide, renal clearance contributes considerably to overall elimination, suggesting that dosage reductions are probably ...
  49. [49]
    Class 1 Antiarrhythmic Drugs (Sodium Channel Blockers) - Lecturio
    Class 1 antiarrhythmic drugs inhibit fast sodium channels in non-nodal myocardial tissue and are subdivided into three categories on this basis.Overview · Class 1A · Class 1B · Class 1C
  50. [50]
    Table: Antiarrhythmic Drugs (Vaughan Williams Classification)
    Antiarrhythmic Drugs (Vaughan Williams Classification) Class Ia Uses: APB and VPB suppression, SVT and VT suppression, AF or atrial flutter, and VF suppression.
  51. [51]
    Comparison of the effect of class IA antiarrhythmic drugs ... - PubMed
    Although quinidine, disopyramide, and procainamide are all classified as Class IA antiarrhythmics, these drugs had different effects on various potassium ...Missing: properties | Show results with:properties
  52. [52]
    2023 ACC/AHA/ACCP/HRS Guideline for the Diagnosis and ...
    Nov 30, 2023 · The 2023 ACC/AHA/ACCP/HRS Guideline for the Diagnosis and Management of Atrial Fibrillation provides recommendations to guide clinicians in the treatment of ...Missing: Ia | Show results with:Ia
  53. [53]
    Antiarrhythmic agents: modulated receptor applications.
    time constants of recovery (tau c 10 msec; figure 3, left) could have little effect on their own; they block the sodium channel for too little time.Missing: Ib | Show results with:Ib
  54. [54]
    Reverse use dependence of antiarrhythmic class Ia, Ib, and Ic
    This phenomenon is called "reverse use dependence." We examined the Ia, Ib, and Ic effects of different Class I drugs on the APD under normal and fast ...
  55. [55]
    Antiarrhythmic drugs - Knowledge @ AMBOSS
    Jan 31, 2025 · Antiarrhythmic drugs are used to prevent recurrent arrhythmias and restore sinus rhythm in patients with cardiac arrhythmias.
  56. [56]
    Mexiletine - StatPearls - NCBI - NIH
    Mexiletine is a sodium channel blocker and further classified as a Class 1B antiarrhythmic in the Vaughan-Williams classification scheme of antiarrhythmic drugs ...
  57. [57]
    Lidocaine CV, Lidopen - Medscape Reference
    1-1.5 mg/kg slow IV bolus over 2-3 minutes. May repeat doses of 0.5-0.75 mg/kg in 5-10 minutes up to 3 mg/kg total if refractory VF or pulseless VT.
  58. [58]
    Lidocaine as an anti‐arrhythmic drug: Are there any indications left?
    Oct 2, 2023 · It increases the ventricular fibrillatory threshold and can interrupt life ... The average half‐life of lidocaine is about 8 to 17 min for the ...
  59. [59]
    Antiarrhythmic Uses of Lidocaine - The Cardiology Advisor
    Mar 14, 2024 · When used to terminate VT, lidocaine should be given as an IV bolus of 1 mg/kg, followed by 1 to 1.5 mg/kg given at a rate of 1 to 3 mg per ...
  60. [60]
    Part 6: Advanced Cardiovascular Life Support | Circulation
    In fact, lidocaine is not an effective or appropriate treatment for SVT. Evidence does not support the use of lidocaine to discriminate between perfusing VT and ...
  61. [61]
    Sodium channel-blocking properties of flecainide, a class IC ...
    ... recovery process of Vmax, whose resultant recovery time constant was 19.9 +/- 1.2 s (n = 6) (time dependence). These flecainide-induced time-dependent ...Missing: tau | Show results with:tau
  62. [62]
    Flecainide Dosage Guide + Max Dose, Adjustments - Drugs.com
    Jul 25, 2025 · Usual Adult Dose for Atrial Fibrillation ... Initial dose: 50 mg orally every 12 hours. Maintenance dose: May be increased in increments of 50 mg ...
  63. [63]
    Propafenone Dosage Guide + Max Dose, Adjustments - Drugs.com
    Jan 29, 2025 · Usual Adult Dose for Atrial Fibrillation: Immediate-release Tablets: Initial dose: 150 mg orally every 8 hours; may increase dose after at least 3 to 4 days to ...
  64. [64]
    Propafenone Therapy and CYP2D6 Genotype - NCBI - NIH
    Apr 4, 2017 · In individuals who lack CYP2D6 activity, metabolism of propafenone is slower, so the 5-hydroxy metabolite is not formed or is formed at very ...Introduction · Drug: Propafenone · Gene: CYP2D6 · The CYP2D6 Gene...
  65. [65]
    Antiarrhythmic Agent - an overview | ScienceDirect Topics
    Drugs with class IC actions are the more potent agents, flecainide and propafenone. Because these are potent sodium channel inhibitors, slowing conduction ...
  66. [66]
    The many NOs to the use of Class IC antiarrhythmics - NIH
    Nov 12, 2022 · Class IC antiarrhythmic drugs (AADs) perform their antiarrhythmic action mainly by blocking rapid sodium channels, thereby slowing down the ...
  67. [67]
    [PDF] 2024 ESC Guidelines for the management of atrial fibrillation ...
    Sep 12, 2024 · Class IC antiarrhythmics (flecainide more so than propafenone) are superior regarding conversion rates within 12 h, while amiodarone.
  68. [68]
    Mortality and Morbidity in Patients Receiving Encainide, Flecainide ...
    Mar 21, 1991 · The CAST study has demonstrated that the use of encainide or flecainide to treat asymptomatic or mildly symptomatic ventricular arrhythmias in ...
  69. [69]
    Topical, Local, and Regional Anesthesia and Anesthetics - NCBI - NIH
    Feb 21, 2025 · Local anesthetics are classified into 2 major chemical families—amino amides and amino esters—each with distinct metabolic pathways.
  70. [70]
    Local Anesthetics: Review of Pharmacological Considerations - PMC
    Local anesthetics interrupt neural conduction by inhibiting the influx of sodium ions through channels or ionophores within neuronal membranes. Normally these ...
  71. [71]
    Essentials of Local Anesthetic Pharmacology - PMC - PubMed Central
    In these situations, bupivacaine (pKa 8.1) would be least effective and mepivacaine (pKa 7.6) would be most likely to provide effective anesthesia (Figure 2).
  72. [72]
    Bupivacaine - StatPearls - NCBI Bookshelf - NIH
    Aug 17, 2023 · Bupivacaine is a potent local anesthetic with unique characteristics from the amide group of local anesthetics, first discovered in 1957.Missing: lidocaine | Show results with:lidocaine
  73. [73]
    Regional Anesthetic Blocks - StatPearls - NCBI Bookshelf - NIH
    A regional block is a specific anesthetic technique that inhibits nerve transmission to avoid or relieve pain.
  74. [74]
    Ultrasound Guided Peripheral Nerve Block Workshop: How to Take ...
    Oct 31, 2025 · This in turn allows for overall reduction in dose needed for the anesthetic and lowers the risk of anesthetic overdose.6. However, USGPNBs ...
  75. [75]
    Voltage-dependent blockade by bupivacaine of cardiac sodium ...
    Nav1.5 is the dominant isoform of VGSCs expressed in cardiac myocytes, and its dysfunction may be the cause of bupivacaine-triggered arrhythmia. Here, we ...
  76. [76]
    A comparative review of epinephrine and phenylephrine as ...
    By inducing vasoconstriction at the injection site, epinephrine reduces blood flow and slows the systemic absorption of the anesthetic agent, thereby allowing ...
  77. [77]
    Sodium Channel Blockers in the Treatment of Epilepsy - PubMed
    Sodium channel blockers have been the mainstay of the pharmacological management of focal and generalised tonic-clonic seizures for more than 70 years.
  78. [78]
    Antiepileptic Drugs: Overview, Mechanism of Action, Sodium ...
    Jun 7, 2024 · The major mechanism of action of ZNS is reduction of neuronal repetitive firing by blocking sodium channels and preventing neurotransmitter ...
  79. [79]
    Cardiac sodium channel inhibition by lamotrigine - PubMed Central
    Lamotrigine, approved for use as an antiseizure medication as well as the treatment of bipolar disorder, inhibits sodium channels in the brain.
  80. [80]
    Lacosamide: A New Approach to Target Voltage-Gated Sodium ...
    Generally, AEDs such as phenytoin, carbamazepine and lamotrigine block sodium channels when activated. In contrast, lacosamide facilitates slow inactivation ...
  81. [81]
    Carbamazepine: Uses, Interactions, Mechanism of Action - DrugBank
    Carbamazepine is an anticonvulsant used to treat various types of seizures and pain resulting from trigeminal neuralgia.
  82. [82]
    CARBAMAZEPINE - Prescriber's Guide – Children and Adolescents
    May 3, 2024 · Maximimal autoinduction generally occurs 2-3 weeks after beginning carbamazepine. Figure 2 Carbamazepine metabolism. Carbamazepine is a strong ...
  83. [83]
    Drug News and FDA Alerts - American Epilepsy Society
    Lacosamide (Motpoly XR) – June 6, 2024 – The FDA approved a new indication for this preparation as adjunctive therapy in the treatment of primary generalized ...
  84. [84]
    Review Sodium channel blockers for the treatment of focal epilepsy
    These multi-mechanism SCBs include topiramate, zonisamide, and cenobamate [19,20]. A summary of SCB mechanisms for seizure control is presented in Fig 2 and ...
  85. [85]
    Carbamazepine-Induced Toxic Effects and HLA-B*1502 Screening ...
    Mar 24, 2011 · A strong association between HLA-B*1502 and carbamazepine-induced SJS–TEN has been found in Asian populations other than the Han Chinese, ...
  86. [86]
    Medications for Arrhythmias - Cardiovascular Disorders
    Class Ia drugs also are used for suppression of atrial or ventricular premature beats. Class Ia drugs may cause torsades de pointes ventricular tachycardia.
  87. [87]
    Pharmacology and Toxicology of Nav1.5-Class 1 anti-arrhythmic drugs
    Although cardiac sodium channel blocking drugs can exert antiarrhythmic actions, they can also provoke life-threatening arrhythmias through a variety of ...
  88. [88]
    Sodium Channel Blocker Toxicity (including tricyclic antidepressants)
    Aug 5, 2025 · This chapter will explore a broad approach to sodium channel blocker toxicity. Much of the data and experience have been obtained from studies of TCA ...Missing: ambiguous | Show results with:ambiguous
  89. [89]
    The Role of Sodium Bicarbonate in the Management of Some Toxic ...
    Aug 8, 2017 · Metabolic acidosis that occurs in cases of hemodynamic deterioration potentiates the sodium blocking activity of TCA medications by increasing ...
  90. [90]
    Cardiovascular Ion Channel Inhibitor Drug-Drug Interactions with P ...
    Cardiovascular ion channel inhibitors are particularly susceptible to DDIs and ADRs with Pgp because they often have low therapeutic indexes.
  91. [91]
    Flecainide: Current status and perspectives in arrhythmia ...
    The reported rate of this proarrhythmic effect is 3.5% to 5.0% and has been associated with high adrenergic conditions[29].
  92. [92]
    Procainamide - StatPearls - NCBI Bookshelf - NIH
    May 8, 2023 · Procainamide monitoring during therapy of an acute arrhythmia should involve monitoring QRS duration via cardiac monitoring, and the clinician ...Missing: serum | Show results with:serum
  93. [93]
    A Literature Review of the Use of Sodium Bicarbonate for the ...
    It has been used for over half a century to treat toxin-induced sodium channel blockade as evidenced by QRS widening on the electrocardiogram (ECG). The purpose ...
  94. [94]
    The Role of Sodium Bicarbonate in the Management of Some Toxic ...
    Clinically the effects of sodium channel blockade may present as cardiac arrhythmias and hemodynamic instability [29]. Metabolic acidosis that occurs in cases ...
  95. [95]
    Clearance of lidocaine by hemodialysis - PubMed
    Lidocaine-urea extraction ratio averaged 11%. Because of the low therapeutic and even toxic plasma levels, the amount of drug removed by dialysis is negligible.
  96. [96]
    Part 10: Adult and Pediatric Special Circumstances of Resuscitation
    Oct 22, 2025 · In these guidelines, the American Heart Association provides updated guidance for resuscitation of adults and children in cardiac arrest or ...
  97. [97]
    An Update to the American Heart Association Guidelines for ...
    Sep 18, 2023 · Many poisons block cardiac sodium channels with properties similar to Vaughan-Williams class Ia or Ic antidysrhythmics. Sodium channel blocker ...
  98. [98]
    An approach to targeting Nav1.7 for pain sensations - JCI
    Jul 15, 2025 · The Nav1.7 voltage-gated sodium channel is a crucial determinant of nociceptor excitability and a promising target for nonaddictive analgesics.Missing: trials | Show results with:trials
  99. [99]
    Nav1.7 and Nav1.8: Role in the pathophysiology of pain - PMC
    Jun 21, 2019 · The aim of this report is to present what is currently known about the contributions of these sodium channel subtypes in the pathophysiology of neuropathic and ...
  100. [100]
    Structure of the Cardiac Sodium Channel
    Dec 19, 2019 · Voltage-gated sodium channel Nav1.5 generates car- diac action potentials and initiates the heartbeat. Here, we report structures of NaV1.5 ...
  101. [101]
    A novel Nav1.7, Nav1.8, and Nav1.9 blocker, ANP-230, has broad ...
    In summary, ANP-230 has shown a superior safety profile and potential for broad analgesic effects in chronic pain conditions. We propose a strategy applying a ...
  102. [102]
    [PDF] Sodium channels as a new target for pain treatment - Frontiers
    Mar 26, 2025 · 7 is considered a threshold channel that regulates the generation of action potentials and is closely associated with various hereditary pain.
  103. [103]
    Selective Inhibition of Na V 1.8 with VX-548 for Acute Pain
    Aug 2, 2023 · The NaV1.8 voltage-gated sodium channel, expressed in peripheral nociceptive neurons, plays a role in transmitting nociceptive signals.
  104. [104]
    NaV1.8 inhibitor poised to provide opioid-free pain relief - Nature
    Dec 12, 2024 · NaV1.8 inhibitor poised to provide opioid-free pain relief. Vertex's first-in-class suzetrigine could soon usher in a new class of analgesic.Missing: sparing | Show results with:sparing
  105. [105]
    Structural mapping of Nav1.7 antagonists | Nature Communications
    Jun 3, 2023 · Here we report high-resolution cryo-EM structures of human Na v 1.7 treated with drugs and lead compounds with representative chemical backbones at resolutions ...
  106. [106]
    Novel Sulfonamide Derivatives as Nav1.5 Sodium Channel Blockers ...
    Apr 21, 2025 · These compounds preferentially inhibit Nav1.5 in atrial cardiomyocytes, extending the effective refractory period and preventing arrhythmic, ...
  107. [107]
    Challenges and Opportunities for Therapeutics Targeting the ...
    Achieving selectivity over off-target NaV isoforms is a formidable challenge in the development of NaV1.7 inhibitors. Due to the obligatory role of NaVs in the ...
  108. [108]
    Challenges and Opportunities for Therapeutics Targeting ... - PubMed
    Oct 10, 2019 · Compelling evidence from human genetic data, particularly the finding that persons lacking functional NaV1.7 are insensitive to pain, has ...
  109. [109]
    Discordance between preclinical and clinical testing of Na V ... - PAIN
    We reviewed preclinical and clinical reports on the analgesic efficacy of Na V 1.7-selective inhibitors and found critical differences in several factors.
  110. [110]
    Evaluation of a New Riluzole‐Based Compound VA945 on Sodium ...
    Oct 31, 2025 · Riluzole (Rilutek), a derivative of benzothiazole, acts as a neuroprotective agent by inhibiting voltage-dependent sodium (Na+) and delaying ...
  111. [111]
    Overexpressed Na V 1.7 Channels Confer Hyperexcitability to in ...
    Here, we show that the expression of Na V 1.7 channels, linked to pain states, is upregulated in KI primary cultures of trigeminal ganglia (TG).Missing: prophylaxis | Show results with:prophylaxis
  112. [112]
    Exosomal TNF-α mediates voltage-gated Na+ channel 1.6 ... - JCI
    Aug 1, 2024 · In conclusion, we provide evidence suggesting that exosomes released by glioma trigger neuronal hyperexcitability in some patients. This action ...
  113. [113]
    [PDF] Suzetrigine for Acute Pain:Effectiveness and Value - ICER
    Mar 31, 2025 · Suzetrigine, a Non-Opioid NaV1.8 Inhibitor for Treatment of Moderate-to-Severe Acute Pain: Two Phase 3 Randomized Clinical Trials.
  114. [114]
    Treated Patients with ALS - Mitsubishi Tanabe Pharma America
    Aug 19, 2025 · The analysis evaluated patients taking RADICAVA ORS® (edaravone) compared with Pooled Resource Open-Access ALS Clinical Trials (PRO-ACT) ...Missing: channel | Show results with:channel
  115. [115]
    An artificial intelligence accelerated virtual screening platform for ...
    Sep 5, 2024 · Structure-based virtual screening plays a key role in drug discovery by identifying promising compounds for further development and refinement.
  116. [116]
    Sodium channel blockers for pain: New opportunities after Vertex's ...
    Jul 1, 2025 · The success of Vertex's opioid alternative Journavx could aid a group of biotechs that aim to take a similar development path with NaV1.8 and NaV1.7 inhibitors.<|separator|>
  117. [117]
    Sodium Channel Blockers Clinical Trial Pipeline Accelerates as 20+ ...
    Apr 9, 2025 · The sodium channel blocker market is expanding due to the increasing prevalence of neuropathic pain and the demand for more selective treatments ...
  118. [118]
    Safety and efficacy of long-term sodium channel blocker therapy for ...
    Sodium channel blockers play a major role in antiarrhythmic drug therapy based on their effectiveness9 and their low risk of extracardiac side effects.