Small Molecule
C22H19ClN6O2S
845272-21-1
Fenfluramine represents a molecule of profound duality in modern pharmacology, embodying a paradoxical journey from a widely proscribed anorectic agent to a highly valued anticonvulsant for rare and catastrophic pediatric epilepsies. Initially approved for obesity and later gaining notoriety as one half of the "Fen-Phen" combination, its widespread use was abruptly halted in 1997 due to an unacceptable risk of potentially fatal valvular heart disease (VHD) and pulmonary arterial hypertension (PAH). This cardiotoxicity was mechanistically linked to the activation of serotonin 5-HT2B receptors on cardiac valve fibroblasts. For over two decades, Fenfluramine was relegated to the annals of cautionary pharmaceutical tales.
However, a deeper understanding of its complex pharmacology has led to its remarkable resurrection. Fenfluramine exerts its effects through a multi-modal mechanism, acting not only as a potent serotonin releasing agent but also as a direct agonist at specific serotonin receptor subtypes (notably 5-HT1D and 5-HT2C) and as a positive modulator of the sigma-1 (σ1) receptor. This combination of enhanced inhibitory neurotransmission and dampened excitatory signaling provides a powerful, network-stabilizing effect. Recognizing this potential, researchers repurposed Fenfluramine at significantly lower doses for the treatment of intractable seizures.
In 2020, under the brand name Fintepla, Fenfluramine received renewed FDA approval for Dravet syndrome, an indication later expanded to include Lennox-Gastaut syndrome. Its use is now governed by a stringent Risk Evaluation and Mitigation Strategy (REMS) program, mandating rigorous cardiac monitoring to manage its known risks. The story of Fenfluramine is therefore a critical case study in the dynamic and context-dependent nature of a drug's risk-benefit profile. It demonstrates that a risk deemed unacceptable for a common condition like obesity can be justifiable when the benefit—control of life-threatening seizures in a treatment-refractory population—is profound, and the risk itself can be systematically monitored and mitigated.
The foundational identity of Fenfluramine is established through a consistent set of chemical, structural, and physical descriptors cataloged across major chemical and pharmacological databases. As a small molecule drug, its properties dictate its pharmacokinetic behavior and formulation characteristics.
Fenfluramine is a synthetic organic compound belonging to the phenethylamine class and is structurally analogous to amphetamine.[1] Its primary identifiers are its DrugBank Accession Number, DB00574, and its Chemical Abstracts Service (CAS) Registry Number, 458-24-2.[1] The molecule is a racemic mixture, often denoted as (±)-fenfluramine or DL-Fenfluramine.[3] Its systematic IUPAC name is
N-ethyl-1-[3-(trifluoromethyl)phenyl]propan-2-amine.[1] In clinical and commercial formulations, it is frequently utilized as its hydrochloride salt (CAS Number: 404-82-0) to improve stability and solubility.[1] A comprehensive list of synonyms includes Fenfluramina, Fenfluraminum, and N-Ethyl-α-methyl-3-(trifluoromethyl)phenethylamine.[3]
The molecular formula for Fenfluramine is .[1] This corresponds to an average molecular weight of approximately 231.26 g/mol and a precise monoisotopic mass of 231.123484 Da.[3] Its two-dimensional structure is unambiguously represented by standard chemical identifiers, including its canonical SMILES string,
CCNC(C)CC1=CC(=CC=C1)C(F)(F)F, and its InChIKey, DBGIVFWFUFKIQN-UHFFFAOYSA-N.[1] These identifiers are crucial for computational modeling and database cross-referencing.
In its pure form, Fenfluramine is described as a colorless to pale yellow oil that is noted to be hygroscopic.[8] It exhibits a melting point of approximately 170°C and a boiling point of 108-112°C at a pressure of 12 Torr.[8] The compound is slightly soluble in organic solvents such as chloroform and methanol.[8] Its basic nature, conferred by the secondary amine group, is quantified by a pKa value reported in the range of 9.10 to 10.22, which influences its absorption and distribution in physiological environments.[5]
Table 1: Compound Identification and Physicochemical Properties of Fenfluramine
Property | Value | Source(s) |
---|---|---|
DrugBank ID | DB00574 | 3 |
CAS Number | 458-24-2 | 1 |
IUPAC Name | N-ethyl-1-[3-(trifluoromethyl)phenyl]propan-2-amine | 1 |
Molecular Formula | 1 | |
Average Weight | 231.26 g/mol | 8 |
Monoisotopic Mass | 231.123484 Da | 3 |
Physical Form | Colourless to Pale Yellow Oil | 8 |
Melting Point | 170°C | 8 |
Boiling Point | 108-112°C (at 12 Torr) | 8 |
pKa | 9.10 - 10.22 | 5 |
InChIKey | DBGIVFWFUFKIQN-UHFFFAOYSA-N | 1 |
Canonical SMILES | CCNC(C)CC1=CC(=CC=C1)C(F)(F)F | 1 |
The pharmacological activity of Fenfluramine is complex, involving multiple neurotransmitter systems and receptor targets. This multi-modal mechanism is central to understanding both its therapeutic efficacy as an anticonvulsant and its historical profile of adverse effects. Its primary actions converge on the serotonergic system, but also extend to sigma-1 receptor modulation, which together create a potent network-stabilizing effect in the central nervous system.
Fenfluramine is a phenethylamine that is structurally similar to the neurotransmitter serotonin (5-hydroxytryptamine, 5-HT).[3] Its principal pharmacodynamic effect is the profound enhancement of serotonergic neurotransmission, achieved through two distinct but complementary actions.
First, it functions as a potent serotonin releasing agent (SRA). It achieves this by interacting with the sodium-dependent serotonin transporter (SERT), disrupting the vesicular storage of serotonin within presynaptic neurons and reversing the transporter's direction of flow.[11] This action effectively pumps serotonin out of the neuron and into the synaptic cleft, leading to a substantial and sustained increase in extracellular serotonin levels.[12]
Second, beyond its effects on serotonin release, Fenfluramine and its major active metabolite, norfenfluramine, act as direct agonists at multiple 5-HT receptor subtypes. Preclinical and in vitro studies have demonstrated agonist activity at 5-HT1D, 5-HT2A, 5-HT2B, and 5-HT2C receptors.[3] The anticonvulsant effects are thought to be mediated primarily through agonist activity at the 5-HT1D and 5-HT2C receptors, which are located in key brain regions like the neocortex and hippocampus.[12] Increased 5-HT signaling generally enhances GABAergic neurotransmission, increasing the frequency and amplitude of inhibitory postsynaptic currents, which helps to quell neuronal hyperexcitability.[12] Conversely, its agonist activity at the 5-HT2B receptor is the primary mechanism underlying its most severe adverse effect, cardiotoxicity.[11]
A more recently elucidated and equally critical component of Fenfluramine's mechanism is its function as a potent positive modulator of the sigma-1 (σ1) receptor.[11] The σ1 receptor is an intracellular chaperone protein that modulates various signaling pathways and ion channels. Fenfluramine's activity at this receptor is believed to inhibit excitatory signaling pathways, likely involving glutamatergic neurotransmission.[13] This action provides a counterbalance to its enhancement of inhibitory signaling.
The profound efficacy of Fenfluramine in treating severe, genetically-driven epilepsies like Dravet syndrome likely arises from the convergence of these distinct mechanisms. While many antiepileptic drugs operate through a single primary mechanism (e.g., sodium channel blockade, GABA agonism), Fenfluramine engages at least two major, complementary pathways. The serotonergic component enhances global inhibitory tone via GABAergic systems, while the sigma-1 receptor modulation simultaneously dampens excitatory drive. This dual-action approach creates a powerful, synergistic effect that can restore the balance between inhibition and excitation in a severely disrupted neuronal network.[13] The fact that potent selective serotonin reuptake inhibitors (SSRIs) do not demonstrate comparable efficacy in these syndromes suggests that the simple elevation of synaptic serotonin is insufficient. It is the combination of broad serotonergic enhancement with the distinct modulatory effect at the sigma-1 receptor that provides the robust, network-level stabilization required to overcome the profound hyperexcitability characteristic of these catastrophic epilepsies.
Fenfluramine's activity extends modestly to other monoamine systems. Its active metabolite, norfenfluramine, also acts as a norepinephrine releasing agent (NRA).[11] At very high, clinically supra-therapeutic concentrations, norfenfluramine may also function as a dopamine releasing agent (DRA).[11] This dopaminergic activity is thought to contribute to the side effect of appetite reduction but its role, if any, in seizure control remains speculative.[13]
Table 2: Summary of Fenfluramine's Pharmacodynamic Actions
Target | Interaction Type | Key Contributor(s) | Associated Clinical Effect(s) |
---|---|---|---|
Serotonin Transporter (SERT) | Reverses transport, Inhibits reuptake | Fenfluramine, Norfenfluramine | Anticonvulsant, Anorectic |
5-HT1D Receptor | Agonist | Fenfluramine, Norfenfluramine | Anticonvulsant |
5-HT2C Receptor | Agonist | Fenfluramine, Norfenfluramine | Anticonvulsant, Anorectic |
5-HT2B Receptor | Agonist | Norfenfluramine (potent) | Cardiotoxic (VHD, PAH) |
Sigma-1 (σ1) Receptor | Positive Modulator | Fenfluramine | Anticonvulsant |
Norepinephrine Transporter (NET) | Reverses transport (weak) | Norfenfluramine | Minor CNS stimulation |
The clinical use of Fenfluramine is guided by its pharmacokinetic profile, which describes its movement into, through, and out of the body.
Following oral administration, Fenfluramine is well absorbed, with an absolute bioavailability of approximately 68-74%.[3] At steady-state, the time to reach maximum plasma concentration (Tmax) is between four and five hours.[3]
Fenfluramine distributes widely throughout the body, as indicated by its large apparent volume of distribution of 11.9 L/kg in healthy subjects.[3] It is moderately bound to plasma proteins, with a binding fraction of 50% that is independent of plasma drug concentration.[3]
Fenfluramine undergoes extensive metabolism, primarily in the liver.[3] This process is mediated by a broad range of cytochrome P450 (CYP) isoenzymes, including CYP1A2, CYP2B6, CYP2D6, CYP2C9, CYP2C19, and CYP3A4/5.[3] The primary metabolic transformation is N-deethylation, which produces the major active metabolite,
norfenfluramine.[3] Norfenfluramine is itself pharmacologically potent, contributing significantly to both the therapeutic anticonvulsant effects and the adverse cardiovascular effects of the parent drug.[8] Several other minor, inactive metabolites are also formed.[3]
The elimination of Fenfluramine and its metabolites is predominantly renal. Over 90% of an administered dose is excreted in the urine, with less than 5% eliminated in the feces.[3] The extensive nature of its metabolism is evident from the fact that unchanged Fenfluramine and its active metabolite norfenfluramine together account for less than 25% of the total amount recovered in urine.[3] In healthy subjects, Fenfluramine has an elimination half-life of approximately 20 hours and a mean clearance of 24.8 L/h.[3]
The clinical and regulatory history of Fenfluramine is one of the most dramatic in modern medicine, characterized by a cycle of widespread use, catastrophic failure, market withdrawal, and eventual redemption through scientific repurposing. This history is essential for understanding its current, highly constrained clinical application.
Fenfluramine was first developed in the early 1960s and introduced for medical use as an appetite suppressant in France in 1963, followed by its approval in the United States in 1973 under the brand name Pondimin.[11] For nearly two decades, it was used as a single agent for the short-term treatment of obesity, achieving only modest popularity due to its limited efficacy and side effects.[15]
Its profile changed dramatically in the early 1990s with the rise of the off-label combination of fenfluramine and phentermine, which became popularly known as "Fen-Phen".[11] This combination, promoted by a 1992 study, was believed to produce more significant weight loss while mitigating the side effects of each individual component.[16] Despite never receiving FDA approval as a combination therapy, Fen-Phen became a cultural phenomenon and a blockbuster prescription, with the number of prescriptions for its constituent drugs soaring to over 18 million in the U.S. by 1996.[16]
The widespread use of Fen-Phen came to an abrupt end in 1997. In August of that year, researchers at the Mayo Clinic published a report in the New England Journal of Medicine linking the drug combination to a unique and dangerous form of cardiac valvulopathy.[15] This was followed by a cascade of reports to the FDA from physicians across the country, confirming the association with two severe, potentially fatal conditions:
valvular heart disease (VHD), a fibroproliferative thickening of the aortic and mitral valves, and pulmonary arterial hypertension (PAH), a rare but deadly form of high blood pressure in the lungs.[1]
Faced with mounting evidence of serious harm, the FDA took decisive action. On September 15, 1997, the agency requested the voluntary withdrawal of both fenfluramine (Pondimin) and its dextro-enantiomer, dexfenfluramine (Redux), from the U.S. market.[15] The withdrawal triggered one of the largest mass tort litigations in history, with over 50,000 lawsuits filed against the manufacturer, American Home Products (later Wyeth), resulting in legal damages estimated to be as high as $14 billion.[15]
Following its withdrawal, Fenfluramine was largely considered a failed and dangerous drug. However, its potent and unique serotonergic mechanism of action intrigued epilepsy researchers.[1] Small-scale use in Belgium had suggested its potential as an effective anticonvulsant in patients with Dravet syndrome, a severe and drug-resistant form of childhood epilepsy.[21] This prompted a new wave of formal clinical development, this time focusing on rare, catastrophic epilepsies at doses significantly lower than those used for weight loss.[22]
Rigorous phase 3 clinical trials demonstrated that low-dose Fenfluramine produced a clinically meaningful and statistically significant reduction in seizure frequency in patients with Dravet syndrome whose seizures were not controlled by other medications.[24] This compelling efficacy data, coupled with a robust plan for managing the known cardiovascular risks, led to a remarkable regulatory reversal. On June 25, 2020, the FDA granted a new approval for Fenfluramine, now marketed as Fintepla, for the treatment of seizures associated with Dravet syndrome.[1] The approval was later expanded to include another severe epilepsy, Lennox-Gastaut syndrome.[11]
The regulatory history of Fenfluramine serves as a powerful illustration of indication-specific risk-benefit analysis. The same molecule, with the same inherent risk of cardiotoxicity, was judged to have two vastly different safety profiles depending on its intended use. For obesity, a prevalent condition with numerous alternative management strategies, the risk of potentially fatal VHD and PAH was deemed unacceptable when weighed against the benefit of moderate weight loss. In this context, the drug failed. However, for Dravet syndrome—a rare, devastating, and life-threatening condition with high mortality and limited effective treatments—the risk-benefit calculus is inverted. The benefit of a substantial reduction in debilitating seizures is immense. The risk of cardiotoxicity, while still a primary concern, is considered manageable through the use of lower doses and the implementation of a mandatory, stringent cardiac monitoring program. This demonstrates that a drug's "safety" is not an absolute, intrinsic property but is instead a relative concept defined by the specific clinical context: the severity of the disease, the availability of alternatives, the patient population, the dosage, and the risk management systems put in place.
In its modern incarnation, Fenfluramine (Fintepla) is a specialized therapy for specific, severe, and often treatment-resistant forms of epilepsy. Its use is supported by robust clinical trial data and is expanding into other rare epileptic encephalopathies.
Fenfluramine is officially indicated for the treatment of seizures associated with Dravet syndrome and Lennox-Gastaut syndrome in patients aged two years and older.[3] These conditions are developmental and epileptic encephalopathies (DEEs) characterized by multiple seizure types, cognitive impairment, and resistance to standard antiepileptic drugs.
In pivotal clinical trials for Dravet syndrome, Fenfluramine demonstrated profound efficacy. One key study showed that patients receiving a higher dose (0.7 mg/kg/day) experienced a 70% median reduction in monthly convulsive seizure frequency (MCSF) relative to placebo.[24] In this group, 70% of patients achieved a clinically significant response, defined as at least a 50% reduction in MCSF, compared to only 7.7% of patients on placebo.[24] Long-term, open-label extension studies have confirmed the durability of this effect, showing a sustained median reduction in MCSF of approximately 67% from baseline over a period of up to three years.[26]
For Lennox-Gastaut syndrome, a condition characterized by multiple seizure types including atonic "drop" seizures that often lead to injury, Fenfluramine also proved effective. Clinical studies demonstrated a statistically significant reduction in the frequency of monthly drop seizures compared to placebo, addressing a core and highly debilitating feature of the syndrome.[28]
The success of Fenfluramine in Dravet syndrome and LGS has prompted investigation into its utility for other severe, rare epilepsies. The drug is currently the subject of multiple clinical trials for other DEEs.
The emerging efficacy of Fenfluramine across a spectrum of genetically and phenotypically distinct epilepsy syndromes—including a sodium channelopathy (Dravet), a syndrome of heterogeneous etiologies (LGS), and disorders caused by other specific gene mutations (CDD, STXBP1)—points toward a broader therapeutic potential. This pattern suggests that its mechanism of action is not targeted to a single defective protein or pathway but rather to a common downstream consequence of these diverse disorders: profound network-level hyperexcitability. By simultaneously enhancing global inhibitory tone through the serotonergic system and dampening excitatory drive via sigma-1 receptor modulation, Fenfluramine provides a robust, network-stabilizing effect. This may allow it to function as a broad-spectrum antiepileptic agent specifically for the most severe and treatment-refractory DEEs, where single-mechanism drugs have failed.
The clinical use of Fenfluramine is inextricably linked to its significant safety concerns, which necessitate a comprehensive risk management strategy. The primary risks are cardiovascular, but its profile also includes notable CNS and metabolic adverse effects.
The U.S. FDA prescribing information for Fintepla carries a Boxed Warning for valvular heart disease (VHD) and pulmonary arterial hypertension (PAH).[11] This is the highest level of warning issued by the FDA and reflects the same serious, potentially fatal risks that led to the drug's withdrawal from the market in 1997.
The mechanism of this cardiotoxicity is well-understood and is associated with serotonergic drugs that possess agonist activity at the serotonin 5-HT2B receptor.[14] These receptors are highly expressed on fibroblasts within human cardiac valves. Stimulation of these receptors by Fenfluramine, and particularly by its more potent metabolite norfenfluramine, is believed to trigger inappropriate mitogenic signaling, leading to fibroblast proliferation and the deposition of extracellular matrix. This results in a characteristic fibroplastic thickening and retraction of the valve leaflets and chordae tendineae, which can impair valve function and lead to regurgitation.[11] A similar pathological process is observed in carcinoid syndrome, a condition of endogenous serotonin overproduction.[34] While clinical trials for the epilepsy indications at lower doses did not identify any cases of VHD or PAH over a period of up to 3 years, the inherent risk remains the foremost safety concern.[21]
To ensure that the benefits of treatment outweigh the significant cardiovascular risks, Fintepla is available only through a restricted distribution program under a Risk Evaluation and Mitigation Strategy (REMS) called the FINTEPLA REMS.[1] This FDA-mandated program imposes strict controls on the prescribing and dispensing of the medication.
Key requirements of the REMS include [11]:
Beyond the boxed warning, Fenfluramine is associated with a range of other adverse effects, primarily affecting the central nervous and gastrointestinal systems.
The most common adverse reactions observed in clinical trials (incidence ≥10% and greater than placebo) vary slightly between the approved indications.[33]
Table 3: Common Adverse Reactions (≥10% Incidence and > Placebo) in Dravet Syndrome and LGS Clinical Trials
Adverse Reaction | FINTEPLA Incidence (%) in Dravet Syndrome | Placebo Incidence (%) in Dravet Syndrome | FINTEPLA Incidence (%) in LGS | Placebo Incidence (%) in LGS |
---|---|---|---|---|
Decreased Appetite | 49 | 10 | 23 | 8 |
Diarrhea | 31 | 10 | 31 | 10 |
Somnolence/Sedation/Lethargy | 27 | 10 | 25 | 9 |
Fatigue/Malaise/Asthenia | 19 | 5 | 26 | 9 |
Vomiting | 15 | 9 | 23 | 11 |
Constipation | 19 | 7 | N/A | N/A |
Abnormal Echocardiogram* | 18 | 6 | N/A | N/A |
Ataxia/Balance/Gait Disturbance | 15 | 3 | N/A | N/A |
Blood Pressure Increased | 13 | 2 | N/A | N/A |
Drooling/Salivary Hypersecretion | 12 | 2 | N/A | N/A |
Pyrexia (Fever) | 12 | 8 | N/A | N/A |
Decreased Weight | 11 | 2 | N/A | N/A |
Fall | 11 | 7 | N/A | N/A |
Status Epilepticus | 11 | 3 | N/A | N/A |
Note: Abnormal echocardiogram findings in trials were primarily physiologic regurgitation, not VHD.27 Data compiled from.33 |
Fenfluramine is contraindicated in patients with a known hypersensitivity to the drug or its excipients.[32] It is also strictly contraindicated for concomitant use with
monoamine oxidase inhibitors (MAOIs) or within 14 days of discontinuing an MAOI, due to the high risk of precipitating a hypertensive crisis or serotonin syndrome.[32] Caution is advised when prescribing to patients with pre-existing cardiac disease, glaucoma, depression, or significant renal or hepatic impairment.[25]
Fenfluramine's complex metabolism and potent pharmacodynamic effects create a high potential for clinically significant drug-drug interactions. One database identifies 835 known drug interactions, of which 108 are classified as major.[40] These interactions can be broadly categorized as pharmacodynamic (additive effects at the site of action) or pharmacokinetic (alteration of drug metabolism).
These interactions result from the combined or opposing effects of drugs on the body's physiological systems.
These interactions occur when one drug affects the absorption, distribution, metabolism, or excretion of another.
Given the large number of potential interactions and the severity of the patient population being treated (who are often on multiple concomitant medications), a thorough review of a patient's entire medication list is essential before initiating Fintepla therapy.
Table 4: Clinically Significant Drug Interactions with Fenfluramine
Interacting Drug / Class | Mechanism of Interaction | Potential Clinical Outcome | Recommended Management |
---|---|---|---|
Monoamine Oxidase Inhibitors (MAOIs) | Pharmacodynamic (Synergistic Serotonergic Effect) | Serotonin Syndrome, Hypertensive Crisis | Contraindicated. Do not use within 14 days of MAOI administration. |
SSRIs, SNRIs, TCAs, Triptans, St. John's Wort | Pharmacodynamic (Additive Serotonergic Effect) | Increased risk of Serotonin Syndrome | Use with caution. Monitor closely for symptoms of serotonin syndrome. |
Strong CYP1A2 or CYP2D6 Inhibitors (e.g., fluoxetine, paroxetine, bupropion) | Pharmacokinetic (Inhibition of Metabolism) | Increased Fenfluramine exposure and risk of adverse effects | Reduce the maximum daily dosage of Fenfluramine as per prescribing information. |
CNS Depressants (e.g., Alcohol, Benzodiazepines) | Pharmacodynamic (Additive Sedation) | Increased somnolence, sedation, and lethargy; impaired motor skills | Avoid concomitant use of alcohol. Use with caution and monitor for excessive sedation. |
Stiripentol | Pharmacokinetic (Inhibition of Metabolism) | Increased Fenfluramine exposure and risk of adverse effects | Reduce the maximum daily dosage of Fenfluramine as per prescribing information. |
Fenfluramine occupies a unique and instructive position in the pharmacopoeia. Its trajectory—from a popular but dangerous diet drug to a life-altering therapy for rare epilepsies—is a powerful testament to the principles of modern, precision-based medicine and rigorous risk management. The story of Fenfluramine underscores the critical concept that the value and safety of a drug are not immutable properties of the molecule itself, but are instead defined by the specific clinical context of its use. A risk-benefit profile that was decisively negative for obesity became positive for catastrophic, treatment-refractory epilepsy, a shift made possible by a deeper understanding of its multi-modal mechanism, the application of lower doses, and the implementation of a robust regulatory framework.
The re-approval of Fenfluramine as Fintepla, governed by the stringent REMS program, represents a landmark case study in regulatory science. It demonstrates that even a drug with a history of severe toxicity can be successfully and responsibly reintroduced for a population with high unmet medical need, provided its risks are well-characterized and can be systematically managed through mandatory monitoring.
Looking forward, the journey of Fenfluramine is not over. Ongoing clinical trials investigating its efficacy in other developmental and epileptic encephalopathies, such as CDKL5 Deficiency Disorder and Sunflower Syndrome, may further expand its role as a potential broad-spectrum agent for the most severe network-based neurological disorders. The long-term cardiovascular safety data that will continue to be collected through the mandatory REMS program will be invaluable for refining our understanding of its risk profile over many years of treatment. Ultimately, Fenfluramine stands not just as an effective anticonvulsant, but as a paradigm-shifting example of drug repurposing, highlighting how meticulous scientific inquiry and adaptive regulatory oversight can reclaim a valuable therapeutic agent from a history of failure.
Published at: October 4, 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.