C19H24N2O2
55268-74-1
Cestode infections, Cysticercosis, Liver fluke infection, Trematode infections, Schistosoma infection
Praziquantel is a pyrazino-isoquinoline derivative that stands as a cornerstone of global public health, recognized on the World Health Organization's List of Essential Medicines for its role as a broad-spectrum anthelmintic agent. It is the drug of choice for the treatment of schistosomiasis and a first-line therapy for a wide array of other trematode (fluke) and cestode (tapeworm) infections. Despite over four decades of widespread clinical use, its precise molecular mechanism of action remains to be fully elucidated, though the current consensus centers on its ability to rapidly and catastrophically disrupt calcium homeostasis within the parasite. This leads to spastic paralysis and severe damage to the parasite's outer surface, the tegument, which facilitates clearance by the host's immune system. The commercially available formulation is a racemic mixture, with the anthelmintic activity residing exclusively in the (R)-enantiomer, a fact that has significant implications for drug formulation, patient tolerability, and future development. Praziquantel exhibits a complex pharmacokinetic profile characterized by rapid absorption, extensive first-pass metabolism mediated by the polymorphic cytochrome P450 system, and a profound food effect that can increase bioavailability up to four-fold. This inherent variability, combined with a significant potential for drug-drug interactions, necessitates careful clinical management. The safety profile of praziquantel is largely favorable for most indications, but adverse effects are directly correlated with parasite burden, and treatment of neurocysticercosis presents unique and severe risks related to the host's inflammatory response to dying intracerebral parasites. This monograph provides an exhaustive analysis of praziquantel's chemical properties, pharmacology, pharmacokinetics, clinical applications, and safety profile, synthesizing current knowledge to provide a definitive reference for clinicians and researchers.
This section details the fundamental chemical identity of praziquantel, its physicochemical properties, the critical role of its stereochemistry in its biological activity, and an overview of its synthetic pathways. These chemical characteristics are foundational to understanding its pharmacological behavior and clinical application.
Praziquantel is a synthetic small molecule classified as a pyrazino-isoquinoline derivative.[1] Its systematic International Union of Pure and Applied Chemistry (IUPAC) name is (RS)-2-(Cyclohexylcarbonyl)-1,2,3,6,7,11b-hexahydro-4H-pyrazino[2,1-a]isoquinolin-4-one.[4] The molecular formula is
C19H24N2O2, and its molar mass is consistently reported as approximately 312.41 g/mol.[2] High-resolution mass spectrometry provides a more precise computed molecular weight of 312.183778013 Da.[1]
Physically, praziquantel is a white to nearly white crystalline powder. It is characterized by a distinctly bitter taste, a property that complicates oral administration, particularly in pediatric populations. The compound is also hygroscopic, meaning it readily absorbs moisture from the air.[2] Its melting point is consistently reported within the range of 136–138 °C, with some sources extending the upper limit to 141 °C with decomposition.[1]
The solubility profile of praziquantel is a key determinant of its pharmacokinetics. It is very slightly soluble in water, with reported values around 0.04 g/100 mL or 400 mg/L.[1] In contrast, it is easily soluble in various organic solvents, including chloroform, dimethylsulfoxide (DMSO), and ethanol.[2] This lipophilic nature is reflected in its partition coefficient (LogP) of 2.5, which facilitates its absorption across the gastrointestinal tract and its penetration of biological membranes, including the blood-brain barrier.[1]
Table 1: Key Chemical and Physical Identifiers of Praziquantel
Identifier Type | Value | Source(s) |
---|---|---|
DrugBank ID | DB01058 | 1 |
CAS Number | 55268-74-1 | 1 |
IUPAC Name | (RS)-2-(Cyclohexylcarbonyl)-1,2,3,6,7,11b-hexahydro-4H-pyrazino[2,1-a]isoquinolin-4-one | 4 |
Molecular Formula | C19H24N2O2 | 4 |
Molar Mass | 312.41 g/mol | 4 |
Exact Mass | 312.183778013 Da | 1 |
Physical Description | White to nearly white crystalline powder; bitter taste; hygroscopic | 2 |
Melting Point | 136–138 °C | 1 |
Water Solubility | ~400 mg/L (0.04 g/100mL) | 1 |
LogP | 2.5 | 1 |
ChEMBL ID | CHEMBL976 | 1 |
PubChem CID | 4891 | 4 |
The drug's inherent physical properties present a direct and significant clinical challenge. The pronounced bitter taste is a primary cause of non-compliance, gagging, and vomiting, especially in children who may be unable to swallow the large 600 mg tablets whole.[9] This has led to strict administration guidelines advising against chewing the tablets and, when necessary, crushing them for mixture with food, a practice that must be completed quickly to mitigate taste aversion.[10] Similarly, its poor aqueous solubility necessitates co-administration with food and liquid to ensure adequate dissolution and absorption, a critical factor for therapeutic efficacy.[9] These properties are not minor inconveniences but are central drivers of the need for improved, more patient-friendly formulations.
Praziquantel possesses a chiral center at the 11b position of its hexahydropyrazino-isoquinoline ring system. Consequently, the drug as it is commercially manufactured and clinically used is a racemic mixture, composed of a 1:1 ratio of two enantiomers: (R)-(−)-praziquantel and (S)-(+)-praziquantel.[2]
Crucially, the biological activity of these two enantiomers is not equivalent. Extensive research has demonstrated that the potent anthelmintic effect of praziquantel is almost exclusively attributable to the (R)-enantiomer.[2] The (S)-enantiomer is largely devoid of parasiticidal activity. Furthermore, the (S)-enantiomer is believed to be a major contributor to the drug's adverse effects, including its prominent bitter taste and potential cardiac toxicity.[3]
This stereospecificity represents a profound inefficiency in the current formulation. For every 600 mg tablet administered, only 300 mg consists of the therapeutically active (R)-enantiomer. The remaining 300 mg is essentially inactive ballast that increases the pill burden and contributes to the side effect profile, thereby hindering patient compliance. This is particularly problematic in the context of mass drug administration (MDA) campaigns, where ease of use and tolerability are paramount for success.[15] The potential for a therapeutic agent with an improved efficacy and safety profile has led the World Health Organization and the research community to anticipate the development of an enantiomerically pure (R)-praziquantel formulation.[3] Such a development would not be an incremental improvement but a transformative one, offering the potential for smaller, more palatable tablets, a reduced side-effect profile, and ultimately, better treatment outcomes in the global fight against schistosomiasis and other helminthic diseases.
Praziquantel was first synthesized in 1975, with its development led by E. Merck and Bayer AG, culminating in its market introduction as Cesol in 1980.[3] The early manufacturing processes were complex and presented significant environmental and safety challenges. They often involved lengthy synthetic routes that utilized highly toxic and hazardous chemicals, such as potassium cyanide and heavy metals, and required stringent reaction conditions like high temperatures and pressures.[3]
Over the years, numerous alternative synthetic pathways have been developed to improve efficiency, safety, and yield. One common and established route involves the acylation of the key intermediate 4-oxo-1,2,3,6,7,11b-hexahydro-4H-pyrazino[2,1-a]isoquinoline with cyclohexanoyl chloride in the presence of a base like triethylamine.[16] Other multi-step processes have been described starting from more basic precursors. For example, one pathway begins with the condensation of β-phenylethylamine with chloroacetyl chloride, followed by a series of reactions to build the pyrazino-isoquinoline core, which is then acylated in the final step to yield praziquantel.[16]
A major focus of modern synthetic chemistry has been the development of enantioselective synthesis routes to produce the pure, active (R)-enantiomer. This is a considerable challenge, but successful strategies have been patented. One such innovative process describes a four-step route that begins with an enantiomerically pure starting material, (1R)-2-substituted-1,2,3,4-tetrahydroisoquinoline-1-carboxylic acid. The synthesis proceeds through a sequence of condensation, reduction, acylation, and finally, a ring-closing reaction to yield (R)-praziquantel.[3] This approach is designed to be more cost-effective, environmentally friendly, and scalable for industrial production by avoiding toxic reagents and harsh conditions. The development of such processes is a critical step toward making an enantiopure formulation a clinical reality.
This section examines the biological effects of praziquantel on susceptible parasites. It details the current understanding of its primary mechanism of action, which centers on the disruption of parasite calcium homeostasis, and explores the resulting pharmacodynamic consequences, including paralysis, tegumental damage, and interaction with the host immune system.
Despite its central role in anthelmintic therapy for over four decades, the precise molecular target and mechanism of action of praziquantel have not been definitively elucidated.[1] However, a substantial body of evidence supports a primary mechanism involving the rapid and profound disruption of calcium ion (
Ca2+) homeostasis within susceptible trematodes and cestodes.[4]
The central tenet of this hypothesis is that praziquantel specifically increases the permeability of the parasite's cell membranes to divalent cations, particularly calcium.[4] This effect is most pronounced at the tegument, the unique syncytial outer layer of the parasite that serves as the primary interface with the host environment. The drug's action leads to a massive, uncontrolled influx of extracellular
Ca2+ into the parasite's cells. This effect is critically dependent on the presence of calcium in the external milieu; in calcium-free media, the characteristic effects of praziquantel are significantly attenuated.[18]
The current scientific consensus points to the parasite's voltage-gated calcium channels (Cav channels) as the most probable molecular target of praziquantel.[18] Studies have identified a unique β subunit of these channels that is specific to platyhelminths (flatworms) and structurally distinct from mammalian counterparts. It is hypothesized that this unique subunit may be responsible for conferring sensitivity to praziquantel.[18] However, the exact binding site on the channel complex has not been identified, and direct, conclusive proof of this interaction is still lacking, leaving the hypothesis unconfirmed.[1]
While the calcium disruption hypothesis is the most widely accepted, other potential or complementary mechanisms have been investigated. These include interactions with the parasite's myosin regulatory light chains, which could influence muscle contraction; inhibition of essential nucleoside uptake, such as adenosine; and effects on the actin cytoskeleton.[18] However, the evidence supporting these alternative mechanisms is less robust than that for the central role of calcium dysregulation. The enduring mystery of its precise molecular target represents a significant vulnerability in the global reliance on praziquantel. This knowledge gap hinders the ability to predict or monitor for the emergence of drug resistance and complicates the rational design of next-generation anthelmintics that could exploit the same target.[18]
The pharmacodynamic effects of praziquantel are a direct and dramatic consequence of the massive intracellular calcium influx it induces. These effects manifest as two distinct but concurrent forms of trauma to the parasite: neuromuscular paralysis and severe tegumental damage.
The most immediate and striking effect is the induction of a rapid, sustained contraction of the parasite's somatic musculature, resulting in spastic paralysis.[4] For schistosomes residing in the mesenteric veins, this paralysis causes them to lose their grip on the blood vessel endothelium. For intestinal tapeworms, it causes detachment from the mucosal wall. This physical dislodgement is the first step in the parasite's clearance from the host.
Simultaneously, praziquantel inflicts severe damage upon the parasite's tegument. The drug causes extensive vacuolization, a process where the surface appears to bubble or form blebs, leading to the eventual disintegration and erosion of this critical protective layer.[5] This tegumental disruption is a crucial component of the drug's efficacy. It is not simply a matter of directly killing the parasite; rather, it serves to "unmask" the worm. The damage exposes parasite-specific surface antigens that were previously shielded from the host's immune system. The paralyzed and structurally compromised worm becomes a conspicuous target for host immune cells, leading to its destruction and clearance via processes such as phagocytosis.[4] This dual action—paralyzing the parasite and simultaneously marking it for immune-mediated destruction—underpins the high efficacy of praziquantel. This mechanism also implies that the drug's success is not entirely independent of the host; a competent host immune response is required to complete the process of parasite elimination. This could have implications for treatment outcomes in immunocompromised individuals, where the clearance phase may be less efficient.
The spectrum of praziquantel's activity is very specific. It is highly effective against a broad range of trematodes (flukes) and cestodes (tapeworms).[1] However, it is notably inactive against nematodes (roundworms), such as filariae, indicating that its molecular target is unique to the phylum Platyhelminthes.[19]
The clinical efficacy and safety of praziquantel are profoundly influenced by its pharmacokinetic profile. This section provides a detailed analysis of how the human body absorbs, distributes, metabolizes, and excretes the drug, highlighting the sources of variability that are critical for clinical consideration.
Following oral administration, praziquantel is absorbed rapidly and almost completely from the gastrointestinal tract, with approximately 80% of the dose being absorbed.[4] Despite this high degree of absorption, the systemic bioavailability of the parent drug is relatively low and exhibits considerable inter-individual variability. This is due to an extensive first-pass effect, where a significant portion of the absorbed drug is immediately metabolized in the liver before it can reach systemic circulation.[4]
Peak serum concentrations (Cmax) are typically achieved within 1 to 3 hours after oral administration in a fasted state.[5] A critically important pharmacokinetic feature of praziquantel is the profound impact of food on its absorption. Co-administration of the drug with food, particularly a high-carbohydrate meal, dramatically increases its bioavailability. Studies have shown that the area under the concentration-time curve (AUC), a measure of total drug exposure, can increase by 2.6-fold with a standard meal and up to 4-fold with a high-carbohydrate meal compared to administration in a fasted state.[13] This effect is thought to be mediated by food-induced increases in splanchnic and hepatic blood flow, which allows more of the drug to bypass first-pass metabolism, as well as potential alterations in the activity of metabolizing enzymes.[13] This "food effect" is not a minor consideration but a non-negotiable clinical instruction. Failure to take praziquantel with a meal can result in sub-therapeutic plasma concentrations and a high risk of treatment failure, a factor with enormous implications for the success of MDA campaigns in resource-limited settings where ensuring a meal with every dose can be a major logistical challenge.
Once in systemic circulation, praziquantel is highly bound to plasma proteins, with approximately 80% of the drug bound, almost exclusively to albumin.[13] This high degree of protein binding means that the concentration of free, pharmacologically active drug can be influenced by conditions that affect plasma albumin levels, such as malnutrition or severe liver disease, which are common in populations affected by schistosomiasis.
The drug distributes widely throughout the body. Non-clinical studies have shown that it concentrates particularly in the liver and kidneys.[13] A key feature of its distribution profile is its ability to effectively cross the blood-brain barrier. This property is essential for its therapeutic efficacy in the treatment of neurocysticercosis, the infection of the central nervous system with tapeworm larvae.[13] Praziquantel is also known to be excreted into breast milk, with concentrations reaching approximately one-quarter of those found in the maternal plasma.[13]
Praziquantel undergoes rapid and extensive metabolism, which is the primary driver of its short half-life and low systemic bioavailability. The liver is the principal site of metabolism, which is mediated by the cytochrome P450 (CYP) enzyme system.[13] Several CYP isoenzymes are involved in its biotransformation, with CYP3A4 being a major contributor, along with CYP1A2, CYP2C9, and CYP2C19.[13]
The primary metabolic pathway is hydroxylation, which results in the formation of several, largely inactive, metabolites. The main metabolite identified in humans is 4-hydroxy-praziquantel, which exists as both cis and trans isomers.[20] While most metabolites are considered inactive, there is some evidence to suggest that the
trans-4-OH-praziquantel metabolite may retain a degree of antischistosomal activity.[23] The metabolism of praziquantel is also stereoselective, meaning the (R) and (S) enantiomers can be metabolized at different rates by the CYP enzymes.[13]
The reliance on multiple, polymorphic CYP enzymes creates a "perfect storm" for pharmacokinetic variability. Genetic variations (pharmacogenetics) in enzymes like CYP2C19 can lead to significant differences in metabolic capacity between individuals, resulting in "poor," "intermediate," or "ultra-rapid" metabolizer phenotypes.[22] Studies have shown that an individual's CYP2C19 genotype can significantly affect their plasma praziquantel concentration.[23] This means that treatment failure in some patients may not be due to parasite resistance but rather to the host's own genetic makeup leading to excessively rapid drug clearance. This inherent variability complicates the interpretation of clinical outcomes and underscores the limitations of a "one-size-fits-all" dosing approach.
Praziquantel and its metabolites are eliminated from the body primarily via the kidneys.[4] Following a single oral dose, approximately 70-80% of the dose is excreted in the urine within 24 hours. The elimination is rapid and consists almost exclusively of metabolites; less than 0.1% of the dose is excreted as the unchanged parent drug.[4]
The extensive metabolism and rapid renal clearance result in a very short elimination half-life (t1/2) for the parent praziquantel molecule, which typically ranges from 0.8 to 1.5 hours in adults with normal renal and hepatic function.[4] The metabolites have a slightly longer half-life, on the order of 4 to 5 hours.[4] This short half-life means that achieving a sufficiently high peak concentration after dosing is critical for the drug's parasiticidal effect.
Table 2: Summary of Praziquantel Pharmacokinetic Parameters (ADME)
Parameter | Value / Description | Source(s) |
---|---|---|
Absorption | ||
Oral Absorption | ~80% | 4 |
Systemic Bioavailability | Low and variable due to extensive first-pass metabolism | 4 |
Food Effect | Bioavailability increases 2.6 to 4-fold with food (especially high-carbohydrate) | 13 |
Tmax (Peak Time) | 1–3 hours | 5 |
Distribution | ||
Protein Binding | ~80% (primarily to albumin) | 13 |
CNS Penetration | Crosses the blood-brain barrier | 13 |
Breast Milk | Excreted at ~25% of plasma concentration | 13 |
Metabolism | ||
Site | Liver (extensive) | 13 |
Primary Enzymes | CYP3A4, CYP1A2, CYP2C9, CYP2C19 | 13 |
Major Metabolite | 4-hydroxy-praziquantel (inactive or weakly active) | 20 |
Excretion | ||
Primary Route | Renal (urine) | 4 |
% Excreted in 24h | 70–80% of dose (as metabolites) | 4 |
% Unchanged in Urine | < 0.1% | 4 |
Half-life (Parent) | 0.8–1.5 hours | 4 |
Half-life (Metabolites) | 4–5 hours | 4 |
Praziquantel's broad spectrum of activity against platyhelminths has established it as an indispensable tool in both human and veterinary medicine. This section details its approved clinical indications, its extensive off-label use which has become the standard of care for many infections, and its applications in veterinary practice.
The primary, regulatory-approved indications for praziquantel in humans focus on infections caused by schistosomes and certain liver flukes.
Beyond its formally approved indications, the clinical utility of praziquantel has expanded dramatically based on decades of clinical experience and research. For many of these "off-label" uses, praziquantel is considered the first-line therapy and standard of care by major public health organizations.
Praziquantel is a cornerstone of veterinary parasitology, where it is used extensively to treat and control cestode (tapeworm) infections in a wide range of animals, including companion animals (dogs and cats), livestock, birds, reptiles, and even fish.[4] Its mechanism of action in animals is the same as in humans, causing paralysis and tegumental disruption that leads to the digestion and elimination of the tapeworm.[4]
It is available in various veterinary formulations, including injectable solutions and oral tablets of different strengths tailored for different animal sizes.[33] Common brand names include Droncit for the single-agent product.[4] Praziquantel is also a frequent component of broad-spectrum combination anthelmintic products, such as Drontal, which typically pair it with drugs active against nematodes (like pyrantel pamoate) to provide comprehensive control of common intestinal parasites in a single dose.[4] Specific veterinary indications include the treatment of salmon poisoning disease in dogs, which involves a rickettsial organism carried by a fluke parasite.[4]
Effective and safe use of praziquantel requires precise dosing based on the specific indication and patient weight, along with strict adherence to administration guidelines. This section synthesizes the recommended dosing regimens and protocols for various patient populations.
The dosing of praziquantel is almost universally calculated based on the patient's body weight in kilograms (mg/kg).[9] The therapeutic approach shows a stark contrast between the short-course, lower-dose regimens for vascular and intestinal parasites and the prolonged, high-dose therapy required for tissue-embedded larval cysts.
Table 3: Summary of Dosing Regimens for Major Indications
Indication | Patient Population (Age) | Recommended Dosage (mg/kg) | Frequency & Duration | Key Notes | Source(s) |
---|---|---|---|---|---|
Schistosomiasis | Adult & Child ≥1 yr | 20 mg/kg | Three times a day for 1 day (4–6 hours apart) | Some guidelines allow 40 mg/kg single or divided dose. | 9 |
Liver Flukes (Clonorchis, Opisthorchis) | Adult & Child ≥1 yr | 25 mg/kg | Three times a day for 1 day (4–6 hours apart) | Treatment may be extended to 2 days for some fluke types. | 9 |
Tapeworms (Taeniasis) (T. saginata, T. solium) | Adult & Child ≥1 yr | 5–10 mg/kg | Single dose | Highly effective for intestinal adult worms. | 27 |
Dwarf Tapeworm (H. nana) | Adult & Child ≥1 yr | 25 mg/kg | Single dose | Higher dose needed to kill cysticercoids in intestinal villi. | 10 |
Neurocysticercosis | Adult & Child ≥1 yr | 50–100 mg/kg/day (divided into 3 doses) | For 14–30 days | Requires hospitalization and co-administration of corticosteroids. | 10 |
This vast difference in total cumulative dose—which can be hundreds of times greater for neurocysticercosis compared to an intestinal tapeworm—is dictated by the parasite's location and life stage. Intraluminal parasites are readily exposed to the drug, whereas tissue-sequestered cysts require prolonged, high-concentration exposure for eradication. This highlights that "praziquantel treatment" is not a single concept; the therapeutic intensity, duration, and associated risks are fundamentally different depending on the diagnosis.
Proper administration is critical for maximizing efficacy and minimizing adverse effects.
This section provides a comprehensive analysis of the safety profile of praziquantel, detailing its common and severe adverse effects, absolute contraindications, and necessary precautions. A crucial aspect of its safety profile is the distinction between direct drug toxicity and the host-mediated inflammatory responses triggered by its therapeutic action.
The tolerability of praziquantel is highly dependent on the host-parasite context. The frequency and severity of adverse effects are directly correlated with the patient's parasite burden; individuals with heavier infections tend to experience more frequent and more pronounced reactions.[4] This observation is a key indicator that many "side effects" are not a result of direct drug toxicity but are rather a consequence of the host's immune system reacting to the sudden death and degradation of a large number of parasites.
Specific situations exist where the use of praziquantel is either absolutely contraindicated or requires significant precautions.
Praziquantel's heavy reliance on the cytochrome P450 metabolic pathway makes it susceptible to a number of clinically significant drug-drug and drug-food interactions. These interactions can either decrease its efficacy to the point of therapeutic failure or increase its concentration to potentially toxic levels.
The majority of significant interactions are pharmacokinetic in nature and are mediated by the induction or inhibition of CYP enzymes, particularly CYP3A4.[13] The list of interacting drugs reads like a catalog of treatments for major global infectious diseases—tuberculosis, HIV, and malaria—that are often co-endemic with schistosomiasis. This creates a high potential for complex clinical scenarios where the treatment for one disease can directly compromise the treatment for another, necessitating an integrated and vigilant approach to patient care.
Table 4: Clinically Significant Drug Interactions with Praziquantel
Interacting Drug/Class | Effect on Praziquantel Plasma Concentration | Clinical Significance & Management Recommendation | Source(s) |
---|---|---|---|
Strong CYP3A4 Inducers | |||
Rifampin | ↓↓↓ (Drastically Reduced) | Contraindicated. Leads to sub-therapeutic levels and certain treatment failure. Discontinue rifampin 4 weeks prior to praziquantel. | 9 |
Carbamazepine, Phenytoin, Phenobarbital | ↓↓ (Significantly Reduced) | Avoid or use with extreme caution. May cause treatment failure. Consider alternative anthelmintic or antiepileptic agents. | 19 |
Dexamethasone | ↓ (Reduced) | Use with caution. May reduce praziquantel levels. Note: High-dose corticosteroids are required for neurocysticercosis, a situation that requires careful management. | 19 |
Efavirenz | ↓ (Reduced) | Avoid or use alternative. May reduce praziquantel efficacy. | 36 |
CYP3A4 Inhibitors | |||
Cimetidine | ↑ (Increased) | Use with caution/Monitor. May increase risk of adverse effects. | 19 |
Azole Antifungals (e.g., Ketoconazole, Itraconazole) | ↑ (Increased) | Use with caution/Monitor. May increase risk of adverse effects. | 27 |
Macrolide Antibiotics (e.g., Erythromycin, Clarithromycin) | ↑ (Increased) | Use with caution/Monitor. May increase risk of adverse effects. | 27 |
Protease Inhibitors (e.g., Atazanavir) | ↑ (Increased) | Use with caution/Monitor. May increase risk of adverse effects. | 27 |
Other Mechanisms | |||
Chloroquine, Hydroxychloroquine | ↓ (Reduced) | Monitor for efficacy. May reduce praziquantel bioavailability. Mechanism is unclear. | 19 |
Albendazole | (Praziquantel ↑ Albendazole levels) | Minor/Significance Unknown. This interaction may be clinically neutral or potentially beneficial. | 27 |
This section outlines the regulatory history of praziquantel, including its approval by the U.S. Food and Drug Administration (FDA), and describes the formulations available for both human and veterinary use.
The original brand name formulation of praziquantel, Biltricide, manufactured by Bayer HealthCare Pharmaceuticals, received its initial New Drug Application (NDA) approval from the U.S. FDA on December 29, 1982.[34] This approval marked a major advancement in the treatment of schistosomiasis and other fluke infections.
Since the expiration of its patent protection, multiple manufacturers have received approval for generic versions of the drug through the Abbreviated New Drug Application (ANDA) pathway. The FDA has determined these generic 600 mg praziquantel tablets to be bioequivalent and, therefore, therapeutically equivalent to the reference listed drug, Biltricide.[34]
The official prescribing information for praziquantel has undergone several revisions over the years to reflect accumulating clinical data and safety information. A notable update was the addition of the contraindication for concurrent use with rifampin, formally acknowledging the critical drug-drug interaction that can lead to therapeutic failure.[44]
Praziquantel remains an indispensable drug in the global armamentarium against parasitic diseases. For over four decades, it has served as a highly effective, low-cost, and generally safe broad-spectrum anthelmintic for treating schistosomiasis, liver fluke infections, and a wide array of cestode infections in millions of people worldwide. Its clinical profile is defined by a complex and fascinating interplay between the drug, the parasite, and the host. Efficacy is driven by a unique pharmacodynamic action that paralyzes the parasite and critically relies on the host's immune system for clearance. This efficacy, however, is subject to the host's highly variable metabolic system, creating a pharmacokinetic profile where factors such as food intake, liver function, drug interactions, and host genetics can profoundly influence clinical outcomes.
Despite its long and successful history, significant knowledge gaps and challenges remain. The most pressing unresolved question is the precise molecular mechanism of action of praziquantel. The inability to definitively identify its molecular target after decades of research is a major scientific and public health vulnerability. Elucidating this mechanism is paramount for understanding and monitoring potential parasite resistance and for the rational design of next-generation drugs that can overcome it.
Looking forward, the most impactful and achievable advancement in praziquantel therapy lies in formulation development. The current racemic mixture represents a significant inefficiency, with half of each dose consisting of an inactive enantiomer that contributes to the pill burden and adverse effect profile. The development, approval, and widespread deployment of an enantiomerically pure (R)-praziquantel formulation is a critical goal. This would offer the potential for smaller, more palatable tablets, improved tolerability, and enhanced patient compliance, particularly in children. Alongside this, the development of a stable, taste-masked pediatric formulation, such as an oral suspension or granules, remains a high priority to address the significant challenges of administering the large, bitter tablets to young children in mass drug administration campaigns.
While praziquantel will undoubtedly remain the cornerstone of schistosomiasis control for the foreseeable future, the deep understanding of its host-dependent effects and pharmacokinetic variability suggests a need to look beyond a "one-size-fits-all" treatment paradigm. Future research should focus on the pharmacogenetics of praziquantel metabolism to better predict individual responses and identify patients at risk of treatment failure. This knowledge could pave the way for more nuanced or personalized dosing strategies, ensuring that this essential medicine can continue to be used to its maximum effect in the fight against neglected tropical diseases.
Published at: August 27, 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.