Small Molecule
C20H26N2O2
4360-12-7
Symptomatic Supraventricular Tachycardia, Tachycardia, Ectopic Junctional, Ventricular Tachycardia (VT)
Ajmaline is a naturally derived monoterpenoid indole alkaloid, classified as a Class Ia antiarrhythmic agent. First isolated from the roots of Rauwolfia serpentina, it has a long history in both traditional and modern medicine. While historically employed for the therapeutic management of tachyarrhythmias, Ajmaline's contemporary clinical role has evolved significantly. Today, its primary and most critical application is as a provocative diagnostic agent for unmasking Brugada syndrome, a rare but potentially lethal genetic channelopathy.
This report provides a comprehensive analysis of Ajmaline, covering its chemical structure, pharmacological mechanisms, pharmacokinetic profile, clinical applications, and global regulatory status. Its potent pharmacological activity stems from a complex mechanism involving the robust inhibition of voltage-gated sodium channels (Nav1.5), supplemented by significant effects on potassium and calcium currents. This multi-channel blockade, while effective, also underlies its primary safety concern: a risk of proarrhythmia.
A key characteristic of Ajmaline is its very short pharmacodynamic half-life, which makes it a relatively safe option for short, highly monitored diagnostic procedures compared to other, longer-acting agents. However, its metabolism is dependent on the highly polymorphic CYP2D6 enzyme, introducing patient-specific variability in drug response and potential toxicity. Strict clinical protocols, including specific dosing, continuous monitoring, and clear termination criteria, are therefore essential for its safe administration. Globally, Ajmaline occupies a niche regulatory space; it lacks centralized approval from major agencies like the U.S. Food and Drug Administration (FDA) but is available in several countries through national authorizations or special access schemes, underscoring its recognized value as an indispensable tool in cardiac electrophysiology.
A precise understanding of Ajmaline's chemical and physical characteristics is fundamental to appreciating its biological activity and pharmaceutical formulation. This section details its nomenclature, structural features, and core physicochemical properties.
Ajmaline is identified by a variety of names and registry numbers across chemical and pharmaceutical databases, which ensures its unambiguous characterization in research and clinical contexts. Its primary designation is Ajmaline, with the DrugBank Accession Number DB01426 and CAS Registry Number 4360-12-7.[1] It is also known by numerous synonyms and trade names that reflect its global commercial history, including Gilurytmal, Ritmos, Aritmina, Cardiorythmine, Raugalline, Rauwolfine, and Tachmalin.[1]
The following table consolidates its key chemical and physical identifiers.
Table 1: Chemical and Physical Identifiers of Ajmaline
| Identifier | Value | Source(s) | 
|---|---|---|
| DrugBank ID | DB01426 | 2 | 
| CAS Number | 4360-12-7 | [1] | 
| PubChem CID | 441080 | 2 | 
| ChEBI ID | CHEBI:28462 | 2 | 
| IUPAC Name | (1R,9R,10S,12R,13S,14R,16S,18R)-13-ethyl-8-methyl-8,15-diazahexacyclo[14.2.1.0¹,⁹.0²,⁷.0¹⁰,¹⁵.0¹²,¹⁷]nonadeca-2,4,6-triene-14,18-diol | 2 | 
| Molecular Formula | $C_{20}H_{26}N_2O_2$ | 1 | 
| Average Molecular Weight | 326.44 g/mol | [1, 4] | 
| Monoisotopic Mass | 326.199428086 Da | 2 | 
| SMILES | CC[C@H]1[C@@H]2C[C@H]3[C@H]4[C@@]5(C[C@@H](C2[C@H]5O)N3[C@@H]1O)C6=CC=CC=C6N4C | 2 | 
| InChIKey | CJDRUOGAGYHKKD-HEFSZTOGSA-N | 2 | 
| Physical Appearance | White to off-white or yellowish solid crystal powder | [1, 5] | 
| Melting Point | 205–207 °C (anhydride) | [2, 5] | 
| pKa | 8.2 (basic, uncertain) | [5, 6] | 
| LogP | 1.81 | [2, 5] | 
Ajmaline is a complex monoterpenoid indole alkaloid. Its core structure is based on the ajmalan skeleton, which is substituted with hydroxy groups at positions 17 and 21.[2] Chemically, it is also classified as a hemiaminal, a functional group that contributes to its chemical reactivity.[2]
The molecule's intricate three-dimensional architecture is defined by a rigid hexacyclic ring system and eight defined stereocenters, as formally described by its IUPAC name.[2] This stereochemical complexity is critical to its biological function. Potent ion channel modulators often rely on a precise three-dimensional conformation to achieve high-affinity binding within the channel pore. The fused, non-planar ring system of Ajmaline severely restricts its conformational flexibility. This structural rigidity effectively pre-organizes the molecule into a shape that is complementary to the binding site within the inner pore of the Nav1.5 sodium channel. This "lock-and-key" relationship, dictated by its stereochemistry, is a primary determinant of its potent channel-blocking activity and differentiates it from more flexible molecules that may bind with lower affinity or specificity.
Ajmaline presents as a white or yellowish crystalline powder.[5] Its solubility profile is characteristic of many alkaloids: it is only slightly soluble in water, with a reported solubility of 489.7 mg/L at 30 °C, but demonstrates significantly better solubility in various organic solvents, including Dimethyl sulfoxide (DMSO) at concentrations of 20–66 mg/mL, Dimethylformamide (DMF) at 25 mg/mL, ethanol at 10 mg/mL, and chloroform.[5] This lipophilicity, reflected in its LogP value of 1.81, facilitates its passage across cell membranes to reach its intracellular target sites on ion channels.[5]
The molecule exhibits a specific optical rotation of $+144^{\circ}$ when measured in chloroform, confirming its chiral nature.[5] For pharmaceutical use, Ajmaline is stable enough for shipment at ambient temperatures but requires controlled storage conditions for long-term preservation. It should be kept in a dry, dark environment at 0–4 °C for short-term storage (days to weeks) or at -20 °C for long-term storage (months to years) to prevent degradation.[1]
The clinical use of Ajmaline is rooted in a rich history of ethnobotany and natural product chemistry. Its journey from a traditional herbal remedy to a specialized modern pharmaceutical highlights the enduring value of botanical sources in drug discovery.
Ajmaline was first isolated in 1931 by the Pakistani scientist Salimuzzaman Siddiqui from the roots of Rauwolfia serpentina, a plant long used in the traditional medicine of the Indian subcontinent.[3] In a tribute to the heritage of this traditional practice, Siddiqui named the compound "ajmaline" in honor of Hakim Ajmal Khan, one of the most distinguished practitioners of Unani medicine in South Asia.[3] This naming connects the purified, single-molecule drug directly to the centuries-old medicinal system from which its source plant emerged.
Ajmaline is a member of the indole alkaloid family and is found widely dispersed across at least 25 plant genera. However, it is most significantly concentrated within the Apocynaceae family, particularly in species of the genus Rauwolfia (such as R. serpentina and R. vomitoria) and in the Madagascar periwinkle, Catharanthus roseus.[3]
Its natural production is a complex, multi-step enzymatic process. The biosynthesis of this monoterpenoid indole alkaloid begins with two primary precursors: the amino acid tryptophan, which provides the indole core, and the iridoid glucoside secologanin, which forms the terpenoid portion.[3] The pathway involves a series of highly specific enzymatic reactions, including a Pictet-Spengler reaction catalyzed by strictosidine synthase to form the key intermediate, strictosidine. This is followed by a cascade of oxidations, methylations, reductions, and acetylations mediated by enzymes such as sarpagan bridge enzymes, vinorine synthase, and various reductases and transferases to ultimately yield the complex hexacyclic structure of Ajmaline.[3]
The reliance on botanical extraction and the complexity of its biosynthesis present significant challenges for consistent pharmaceutical production. The manufacturing process involves extracting the alkaloid from plant roots followed by extensive chromatographic purification.[5] This method is inherently susceptible to batch-to-batch variability in yield and purity, which can impact the final drug product's potency and safety.[14] These production hurdles, compared to the more controlled and scalable processes of fully synthetic drugs, contribute to Ajmaline's limited global availability and reinforce its status as a specialized, high-value agent rather than a mainstream therapeutic. Furthermore, the low oral bioavailability of natural Ajmaline prompted the development of a semi-synthetic propyl derivative, prajmaline (trade name Neo-gilurythmal), which was designed to improve absorption and provide a more predictable clinical effect, demonstrating a clear path of pharmaceutical optimization originating from a natural product lead.[3]
Ajmaline exerts its profound effects on cardiac electrophysiology through a complex interplay of interactions with multiple ion channels. While primarily classified by its action on sodium channels, its unique clinical profile is defined by its broader, multi-target mechanism.
Ajmaline is formally classified as a Class Ia antiarrhythmic agent under the Vaughan Williams classification system.[1] Drugs in this class are characterized by their moderate blockade of the fast inward sodium current ($I_{Na}$) and their intermediate kinetics of binding to and dissociating from the sodium channel. This action has two principal electrophysiological consequences:
The primary molecular target of Ajmaline is the voltage-gated sodium channel, particularly the cardiac isoform Nav1.5, which is encoded by the SCN5A gene.[8] Ajmaline acts as a potent inhibitor of this channel, physically obstructing the pore and reducing the influx of sodium ions ($Na^+$) during Phase 0 of the cardiac action potential.[1] This direct inhibition of the depolarizing current is the fundamental reason for its ability to slow cardiac conduction.[3]
A defining feature of Ajmaline's pharmacology is that it is not a "pure" sodium channel blocker. Its clinical effects, particularly its efficacy in diagnosing Brugada syndrome, are significantly influenced by its actions on other ion channels.[17]
This "pharmacological impurity" is, paradoxically, the key to its diagnostic power. Brugada syndrome is a complex channelopathy, with genetic variants identified not only in the sodium channel gene SCN5A but also in genes encoding potassium and calcium channels.[19] A drug that only blocked Nav1.5 might not be sufficient to unmask the subtle electrical abnormalities present in all patients. By simultaneously inhibiting $I_{Na}$, $I_{to}$, $I_{Kr}$, and $I_{Ca,L}$, Ajmaline places the entire cardiac cellular system under profound electrical stress. This multi-pronged challenge is highly effective at exaggerating the underlying repolarization abnormalities in the right ventricle, making it a more potent and sensitive diagnostic tool for this specific condition than more selective agents.[20]
The combined effects of Ajmaline's multi-channel blockade produce distinct and measurable changes on the surface electrocardiogram (ECG).
Together, these actions significantly increase the cardiac refractory period, which is the basis for its antiarrhythmic effect in treating re-entrant tachycardias.[3] In the context of Brugada syndrome, this potent and broad-spectrum channel blockade unmasks the characteristic ECG pattern: a "coved" ST-segment elevation of at least 2 mm in one or more right precordial leads (V1-V3), which is diagnostic for the condition.[21]
The disposition of Ajmaline within the body—its absorption, distribution, metabolism, and excretion (ADME)—is characterized by rapid onset, extensive tissue distribution, and a primary reliance on hepatic metabolism, which has significant clinical implications for both efficacy and safety.
For its primary clinical applications, Ajmaline is administered via intravenous (IV) injection. This route ensures 100% bioavailability and a rapid onset of action, which is critical for both acute arrhythmia treatment and the controlled conditions of a diagnostic provocation test.[2] The parent compound is known to have low oral bioavailability, which led to the development of derivatives like prajmaline for oral use.[3]
The metabolism of Ajmaline is complex and involves several biotransformation reactions, including mono- and di-hydroxylation of its aromatic ring, O-methylation, reduction of the C-21 position, and N-oxidation.[17]
The primary enzyme responsible for catalyzing these metabolic transformations is Cytochrome P450 2D6 (CYP2D6).[17] The gene encoding this enzyme is known to be highly polymorphic in the human population, with over 70 different alleles identified, leading to significant inter-individual variability in metabolic capacity. Individuals can be classified into phenotypes ranging from poor metabolizers (PMs) to ultrarapid metabolizers (UMs). This genetic variability has profound clinical importance. A patient who is a CYP2D6 poor metabolizer will clear Ajmaline much more slowly than expected. Following a standard weight-based dose, this individual would be exposed to higher plasma concentrations for a longer duration, significantly increasing the risk of toxicity, proarrhythmia, or a false-positive result during a diagnostic challenge.[17] This highlights a potential role for pharmacogenetic testing prior to an Ajmaline challenge to identify at-risk patients and potentially adjust dosing, although this is not yet standard practice.
The plasma concentration of Ajmaline after an IV bolus follows a triexponential decay curve, reflecting a rapid distribution phase followed by a slower elimination phase.[23]
A critical point of nuance lies in the drug's half-life. While its terminal elimination half-life ($t_{1/2}$), which reflects the time taken to clear the drug from the body, is approximately 7.3 hours, its pharmacodynamic half-life, which reflects the duration of its clinical effect, is much shorter, reported to be around 5–6 minutes.[6] This discrepancy is clinically advantageous. The short duration of action ensures that the period of acute risk during a diagnostic test is brief and manageable, allowing the drug's effects to dissipate quickly once the infusion is stopped. This is a key reason for its preferred status over longer-acting agents like flecainide.[25] However, the much longer elimination half-life, combined with its metabolism by the polymorphic CYP2D6 enzyme, means that the potential for drug accumulation and delayed toxicity exists, particularly in patients with impaired metabolic capacity.
The following table summarizes the key pharmacokinetic parameters of Ajmaline.
Table 2: Key Pharmacokinetic Parameters of Ajmaline
| Parameter | Reported Value | Clinical Significance | Source(s) | 
|---|---|---|---|
| Volume of Distribution ($V_d$) | 4 – 6.17 L/kg | Indicates extensive distribution into tissues, with low concentration in plasma. | 6 | 
| Plasma Protein Binding | 29% – 46% | A large fraction of the drug (54-71%) is unbound and pharmacologically active. | 23 | 
| Total Plasma Clearance (CL) | 9.76 – 9.80 mL/min/kg | The drug is cleared efficiently from the plasma, primarily by the liver. | 6 | 
| Route of Elimination | Primarily hepatic metabolism (>95%); minor renal excretion (<5%) | Clearance is highly dependent on liver function and CYP2D6 enzyme activity. | 23 | 
| Pharmacodynamic Half-life | ~5–6 minutes | The primary clinical and electrophysiological effects are very short-lived, enhancing safety in diagnostic tests. | [24] | 
| Terminal Elimination Half-life ($t_{1/2}$) | ~7.3 hours | Reflects the time to clear the drug from the body; risk of accumulation in poor metabolizers. | 6 | 
Ajmaline's clinical utility has undergone a significant evolution. Originally valued as a therapeutic agent for a range of arrhythmias, its potent but complex pharmacological profile has led to its repositioning as a highly specialized diagnostic tool, particularly for the identification of Brugada syndrome.
The primary contemporary use of intravenous Ajmaline is in the Ajmaline Provocation Test. This test is the gold standard for diagnosing Brugada syndrome in individuals who have a suspicious clinical history (e.g., unexplained syncope, nocturnal agonal respiration, or a family history of sudden cardiac death) but a non-diagnostic or equivocal baseline ECG.[1]
The test is designed to pharmacologically "unmask" the latent electrical substrate of the syndrome. By potently blocking sodium and other ion channels, Ajmaline induces or exaggerates the characteristic ECG pattern of Brugada syndrome: a coved-type ST-segment elevation of $\geq2$ mm in at least one right precordial lead (V1 to V3), often referred to as the "Type 1" pattern.[21] The appearance of this pattern confirms the diagnosis. Ajmaline is often considered superior to other sodium channel blockers, such as flecainide or procainamide, for this purpose due to its shorter half-life, which enhances safety by limiting the duration of proarrhythmic risk, and its reported higher sensitivity in unmasking the diagnostic ECG pattern.[25]
While now used less frequently for therapy, Ajmaline has a documented history of efficacy in the acute management of various supraventricular and ventricular tachyarrhythmias.[2] Its potent sodium channel blocking effects make it effective for the termination of arrhythmias that rely on re-entrant circuits. Historical and ongoing use in some countries includes:
This shift away from routine therapeutic use towards a specialized diagnostic role reflects a broader trend in cardiovascular medicine. As safer, more specific, and orally available antiarrhythmic drugs have been developed, older agents with narrow therapeutic windows and significant proarrhythmic potential like Ajmaline have been reserved for acute, controlled settings where their potent effects provide a unique benefit that outweighs the risks. The risk-benefit calculation favors its use in a short, monitored test to diagnose a life-threatening condition, but is less favorable for chronic therapeutic management where safer alternatives exist.
The clinical use of Ajmaline is supported by a body of evidence from clinical trials and large observational studies. A completed Phase 4 clinical trial (NCT00702117) specifically investigated the utility of Ajmaline in the diagnosis and treatment of cardiac arrhythmias, including atrial fibrillation, sudden unexplained death syndrome (a term often associated with Brugada syndrome), and ventricular tachycardia.[29]
Numerous observational studies have evaluated the safety and efficacy of the Ajmaline challenge. A large retrospective analysis of 677 consecutive patients undergoing the test found a positive result (induction of the Type 1 Brugada pattern) in 39% of cases. The study concluded that the test was safe when conducted according to a strict protocol and helped identify predictors of a positive response, such as a baseline saddleback-type ECG pattern.[27]
Its use has also been studied in pediatric populations. Reports from specialist centers indicate that the Ajmaline provocation test is feasible and generally safe in children when performed by experienced teams in an appropriate setting.[30] However, some data suggest that the risk of inducing ventricular arrhythmias during the test may be higher in certain pediatric cohorts, warranting increased caution.[32]
The administration of Ajmaline, particularly for the provocation test, is governed by strict, standardized protocols designed to maximize diagnostic yield while minimizing the inherent risk of proarrhythmia. The procedure is a controlled pharmacological stress test that demands a high level of clinical vigilance.
The standard protocol for the Ajmaline challenge involves the intravenous administration of the drug in a carefully controlled hospital setting, typically a cardiac catheterization lab or coronary care unit.[26]
The safety of the Ajmaline challenge hinges on continuous monitoring and strict adherence to predefined termination criteria. The procedure is not merely an injection but a dynamic assessment where the primary risk mitigation strategy is the early recognition of warning signs and immediate cessation of the drug infusion.
This highly structured approach underscores the recognized potential for harm. The entire procedure is designed around the principle of a controlled challenge, where the line between a diagnostic outcome and a life-threatening arrhythmia is narrow. The skill of the administering team lies in their ability to titrate the pharmacological stress against the patient's real-time electrophysiological response, knowing precisely when the diagnostic or safety threshold has been crossed.
While a valuable clinical tool, Ajmaline possesses a narrow therapeutic index and a significant potential for adverse effects. Its safety profile is a direct extension of its potent, multi-channel blocking mechanism, necessitating careful patient selection and risk management.
Adverse reactions to Ajmaline range from common, benign sensory effects to rare, life-threatening cardiac events.
The contraindications for Ajmaline use are logically derived from its mechanism of action and are designed to protect patients whose cardiac systems are already electrically or structurally vulnerable. Administering Ajmaline to these patients would subject them to an unacceptable level of risk.
Absolute contraindications include 41:
Particular caution is also warranted in any patient with pre-existing intraventricular conduction disturbances, such as a wide QRS complex at baseline.[15]
The risk of adverse events is significantly increased when Ajmaline is co-administered with other drugs that affect cardiac electrophysiology or its metabolism. The following table outlines the most critical interactions.
Table 3: Clinically Significant Drug Interactions with Ajmaline
| Interacting Drug/Class | Pharmacodynamic/Pharmacokinetic Effect | Clinical Recommendation/Risk | Source(s) | 
|---|---|---|---|
| QTc Prolonging Agents (e.g., certain macrolide and fluoroquinolone antibiotics, antipsychotics, tricyclic antidepressants, azole antifungals) | Additive effect on potassium channel blockade, leading to excessive QT prolongation. | Increased risk of life-threatening Torsades de Pointes. Co-administration should be avoided. | [8, 42] | 
| Other Antiarrhythmic Agents (e.g., Class I, Class III agents like amiodarone, sotalol; beta-blockers like acebutolol) | Additive or synergistic antiarrhythmic and proarrhythmic effects. Can lead to excessive bradycardia, conduction block, or ventricular arrhythmias. | Co-administration is generally contraindicated or requires extreme caution and expert consultation. | [8, 43] | 
| Adenosine | May increase the arrhythmogenic activities of Ajmaline. | Increased risk of arrhythmia induction. Use with caution. | [8] | 
| CNS Depressants (e.g., benzodiazepines like diazepam, barbiturates like phenobarbital) | Potential for additive Central Nervous System (CNS) depression. | May increase the risk or severity of sedation and respiratory depression. Monitor patient closely. | [42] | 
| CYP2D6 Inhibitors (e.g., fluoxetine, paroxetine, bupropion) | Inhibition of Ajmaline's primary metabolic pathway, leading to increased plasma concentrations and prolonged half-life. | Increased risk of toxicity and proarrhythmia. Use with caution or avoid. | 17 | 
Ajmaline occupies a unique and complex position in the global pharmaceutical landscape. Despite its recognized clinical importance, particularly in Europe, it is not a widely approved or marketed drug, and its availability is heterogeneous across different regulatory jurisdictions.
This regulatory pattern positions Ajmaline as a quintessential "orphan diagnostic" agent. Its clinical utility is well-established and endorsed by expert guidelines for a rare but serious condition. However, the limited patient population and the challenges of its botanical sourcing and manufacturing likely make the pursuit of full, centralized marketing authorization in major jurisdictions like the U.S. commercially unviable for pharmaceutical companies. Consequently, health systems rely on alternative regulatory pathways—such as national approvals, special access schemes, and investigational protocols—to ensure that this medically necessary drug remains available to the specialist physicians who need it.
Reflecting its regulatory status, Ajmaline's global availability is limited and concentrated. It is marketed under trade names such as Gilurytmal, Ritmos, and Aritmina in regions where it holds national approval.[3] The market is dominated by a small number of specialized suppliers, with companies like Solvay and Lacer being prominent in certain European markets.[14] For most of the world, access is not through standard retail or wholesale pharmacy channels but is restricted to hospital pharmacies in specialist cardiology centers that have obtained the drug via direct purchase or special importation procedures.[14]
Ajmaline represents a fascinating case study in the lifecycle of a pharmaceutical agent. Originating from a plant used in traditional medicine, it was developed into a potent, multi-target antiarrhythmic drug. Over time, with the advent of safer and more specific therapies, its role has pivoted from broad therapeutic application to a highly specialized, indispensable diagnostic tool. Its primary contemporary value lies in its ability to unmask Brugada syndrome, a diagnosis that carries profound implications for patient management and the prevention of sudden cardiac death.
Its efficacy in this role is a direct consequence of its complex pharmacology—a potent blockade of sodium channels combined with significant effects on potassium and calcium currents. This same mechanism, however, is the source of its considerable proarrhythmic risk. Consequently, its clinical use is tenable only within the confines of a strict, meticulously monitored protocol where the line between diagnosis and danger is managed by expert vigilance. Its unique pharmacokinetic profile, with a short duration of action but a long elimination half-life dependent on the polymorphic CYP2D6 enzyme, adds another layer of complexity to its risk-benefit assessment. Ajmaline's journey underscores a critical principle in modern medicine: even as new drugs are developed, older, potent agents can retain immense value when repurposed for niche applications where their unique properties provide a benefit that cannot be replicated by newer alternatives.
While Ajmaline's current role is well-defined, several avenues for future research could enhance its safety and clinical utility.
Published at: October 30, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.