C16H13N3O3
31431-39-7
Ancylostoma caninum infection, Ancylostoma duodenale infection, Ascaris lumbricoides infection, Capillariasis, Enterobius vermicularis infection, Filariasis, Giardiasis, Necatoriasis due to necator americanus, Strongyloides Stercoralis Infection, Taenia solium infection, Toxocariasis, Whipworm infection
This section establishes the fundamental chemical and physical identity of mebendazole, providing the nomenclature, identifiers, and properties that underpin its formulation, pharmacokinetic behavior, and mechanism of action.
Mebendazole is a synthetic small molecule compound recognized across numerous chemical and pharmacological databases by a standardized set of identifiers.[1] Its formal chemical name according to the International Union of Pure and Applied Chemistry (IUPAC) is Methyl (5-benzoyl-1H-benzimidazol-2-yl)carbamate, though it is also cited as methyl N-(6-benzoyl-1H-benzimidazol-2-yl)carbamate.[1] The compound is uniquely identified by its Chemical Abstracts Service (CAS) Registry Number, 31431-39-7.[1]
In pharmaceutical and biomedical research, it is cataloged under DrugBank Accession Number DB00643.[1] Other key identifiers include PubChem Compound ID 4030, UNII 81G6I5V05I, and ChEMBL ID CHEMBL685.[1] During its development and in research contexts, it has been referred to by codes such as R 17635 and NSC 184849, and by synonyms including 5-Benzoyl-2-benzimidazolecarbamic acid methyl ester.[3]
Commercially, mebendazole is marketed globally under a wide array of brand names. The most prominent include Vermox, Ovex, and Emverm.[1] Other international brand names include Pantelmin, Lomper (Spain), Surfont (Germany), Bendax (Egypt), Mebex (India), and Antiox (Philippines), reflecting its worldwide use.[5]
Mebendazole is a synthetic derivative of the benzimidazole class of compounds, which is characterized by a fused benzene and imidazole ring system.[2] Its molecular formula is
C16H13N3O3.[1] Structurally, it is a carbamate ester and an aromatic ketone.[2] The core structure is a methyl 1H-benzimidazol-2-ylcarbamate, which is substituted at the 5-position of the benzimidazole ring with a benzoyl group.[2] This specific chemical architecture is fundamental to its biological activity and places it within the same therapeutic family as other broad-spectrum anthelmintics like albendazole and fenbendazole.
The therapeutic utility and pharmacokinetic profile of mebendazole are directly governed by its distinct physicochemical properties. With a molar mass of approximately 295.298 g·mol⁻¹, it exists physically as a white to slightly yellow or off-white amorphous powder or crystal.[1] It is noted to have a not unpleasant or even pleasant taste, which is advantageous for its oral chewable formulations.[2]
A defining and clinically critical characteristic of mebendazole is its extremely low aqueous solubility. It is classified as practically insoluble in water, as well as in common organic solvents like alcohol and methylene chloride.[2] Quantitative measurements confirm this, with reported solubility values around 0.035 mg/mL (or 35.4 mg/L) at 25 °C.[11] This poor solubility is the primary determinant of its pharmacokinetic behavior. It leads to low and incomplete absorption from the gastrointestinal tract, resulting in a bioavailability of only 2-20%.[1] This characteristic creates a therapeutic paradox: it is highly advantageous for treating intestinal parasites, as it concentrates the drug locally in the gut lumen while minimizing systemic exposure and side effects.[1] However, this same property presents a significant challenge when systemic drug levels are required, such as in the treatment of systemic parasitic diseases or for its repurposed use in oncology.[13] This limitation necessitates clinical strategies to enhance absorption, such as co-administration with high-fat meals or the development of advanced drug delivery systems.[13]
Mebendazole exhibits polymorphism, existing in at least three different crystalline forms: A, B, and C.[18] These polymorphs possess different physical properties, including solubility. In an acidic medium (0.1 M HCl), the solubility follows the order B > C > A, a factor that is highly relevant for pharmaceutical formulation and dissolution kinetics.[18] Other key properties include a high melting point of approximately 288.5 °C and pKa values of 3.43 and 9.93 (uncertain).[2] Based on these properties, mebendazole is categorized under the Biopharmaceutical Classification System (BCS) as Class II or IV, signifying low solubility and variable permeability, which aligns with its observed pharmacokinetic challenges.[11]
Table 1: Key Identifiers and Physicochemical Properties of Mebendazole
Property | Value | Source(s) |
---|---|---|
IUPAC Name | Methyl (5-benzoyl-1H-benzimidazol-2-yl)carbamate | 1 |
CAS Number | 31431-39-7 | 1 |
DrugBank ID | DB00643 | 1 |
PubChem CID | 4030 | 1 |
UNII | 81G6I5V05I | 1 |
Molecular Formula | C16H13N3O3 | 1 |
Molar Mass | 295.298 g·mol⁻¹ | 1 |
Physical Appearance | White to slightly yellow crystalline powder | 2 |
Melting Point | 288.5 °C | 2 |
Aqueous Solubility | Practically insoluble (~0.035 mg/mL at 25 °C) | 11 |
BCS Class | Class II / IV | 11 |
Polymorphic Forms | A, B, C | 18 |
The pharmacological activity of mebendazole is defined by its ability to disrupt fundamental cellular processes through a specific molecular interaction. This core mechanism is the unifying principle that explains its efficacy in two disparate therapeutic fields: parasitology and oncology. By functioning as a potent tubulin modulator, mebendazole effectively targets the cytoskeletal integrity of both parasitic helminths and rapidly dividing mammalian cancer cells.
Mebendazole's principal anthelmintic effect is achieved through the targeted disruption of microtubule structures within the intestinal cells of parasitic worms.[1] The drug exhibits a high affinity for and selectively binds to the colchicine-binding site on the β-tubulin subunit of the parasite's tubulin protein.[1] This binding event physically obstructs the polymerization of tubulin dimers into the long, filamentous polymers that form cytoplasmic microtubules.[1]
The resulting loss of the microtubular cytoskeleton triggers a cascade of catastrophic failures in the parasite's cellular functions. Most critically, it incapacitates the absorptive intestinal cells, blocking the uptake of glucose and other essential nutrients from the host's gut.[1] This leads to a rapid depletion of the worm's glycogen stores and a severe reduction in the production of adenosine triphosphate (ATP), the primary energy currency of the cell.[15] The ensuing energy crisis results in the gradual immobilization and paralysis of the helminth, ultimately leading to its death.[1] The dead or debilitated parasites are then expelled from the host's gastrointestinal tract via normal peristalsis over a period of several days.[1] In addition to its effects on adult worms, mebendazole also demonstrates ovicidal activity against the eggs of certain species, such as
Ascaris lumbricoides and Trichuris trichiura, contributing to its overall therapeutic efficacy.[13]
The clinical success and favorable safety profile of mebendazole as an anthelmintic agent are fundamentally rooted in its selective toxicity. The drug's binding affinity for helminthic β-tubulin is significantly higher than its affinity for the analogous protein in mammals.[15] This differential binding means that at standard therapeutic concentrations, mebendazole is profoundly disruptive to the parasite's cellular machinery while exerting only minimal effects on the host's cells. This selectivity is the key to achieving a wide therapeutic window, allowing for effective parasite eradication with a low incidence of host toxicity.[15]
Despite its selectivity for parasitic tubulin, mebendazole does interact with and inhibit the polymerization of mammalian tubulin, particularly at the higher concentrations employed in oncological research.[1] This interaction forms the basis of its anti-cancer activity. The primary target in mammalian cells is the mitotic spindle, a highly dynamic microtubule-based structure that is essential for the segregation of chromosomes during cell division (mitosis).[1]
By disrupting the formation and function of the mitotic spindle, mebendazole causes dividing cells to arrest in the M-phase of the cell cycle. This prolonged mitotic arrest is an unsustainable state for the cell and triggers the intrinsic pathway of programmed cell death, or apoptosis.[1] Mechanistically, this has been shown to be mediated through the dephosphorylation of Bcl-2, an anti-apoptotic protein. The inactivation of Bcl-2 allows the pro-apoptotic protein Bax to form dimers, which then initiate the downstream caspase cascade that executes cell death.[1] This specific pathway has been demonstrated to be effective even in chemoresistant melanoma cells.[4]
The mechanism of inducing mitotic arrest via tubulin disruption is a well-established and validated strategy in cancer chemotherapy. It is the same fundamental mechanism employed by classic chemotherapeutic drug classes such as the taxanes (e.g., paclitaxel) and the vinca alkaloids (e.g., vincristine).[21] Therefore, mebendazole is not merely an anthelmintic with incidental anti-cancer properties; it is a legitimate microtubule-destabilizing agent and mitotic inhibitor, providing a strong scientific rationale for its repurposing in oncology.
The clinical behavior of mebendazole is dictated by its distinct pharmacokinetic profile. Characterized by poor absorption but excellent tissue penetration of the absorbed fraction, its journey through the body explains both its efficacy in localized intestinal infections and its potential for systemic activity at higher doses.
Following oral administration, mebendazole is poorly and erratically absorbed from the gastrointestinal tract.[1] Its systemic bioavailability is very low, with estimates ranging from 2-10% to 5-20% of the administered dose.[1] This poor absorption is a direct consequence of its low aqueous solubility, which limits its dissolution in the gut lumen, combined with extensive presystemic (first-pass) metabolism in the gut wall and liver.[13]
As a result, systemic plasma concentrations achieved with standard anthelmintic doses are minimal. For instance, following a regimen of 100 mg twice daily for three days, peak plasma levels of mebendazole and its primary metabolite typically do not exceed 0.03 µg/mL and 0.09 µg/mL, respectively.[23] A significant food effect has been observed; co-administration with a high-fat meal can substantially increase the drug's absorption and bioavailability.[13] This is a critical clinical consideration for any indication where achieving systemic therapeutic concentrations is the goal.
Mebendazole exhibits an apparent pharmacokinetic contradiction: despite being poorly absorbed, the small fraction that does enter the systemic circulation distributes exceptionally well into tissues. Once in the bloodstream, it is highly bound to plasma proteins, with a binding fraction of 90-95%.[1]
The drug has a large apparent volume of distribution, estimated at 1 to 2 L/kg, which indicates that it does not remain confined to the bloodstream but penetrates extensively into extravascular tissues.[11] This suggests that while the total quantity of drug entering the body is small, its physicochemical properties—notably its lipophilicity and low molecular weight—make it highly efficient at partitioning from the plasma into peripheral tissues.
Crucially, these properties also allow mebendazole to cross the blood-brain barrier.[13] This capacity for central nervous system penetration is clinically validated by its effectiveness in treating cerebral echinococcosis and is the cornerstone of the rationale for investigating its use in neuro-oncology.[13] This demonstrates that "low bioavailability" does not preclude systemic or CNS activity; even low plasma concentrations can translate into therapeutically meaningful drug levels at the target site, particularly when the high doses used in oncology are administered.
The absorbed fraction of mebendazole is rapidly and extensively metabolized, primarily by cytochrome P450 enzymes in the liver.[14] The principal metabolic pathway is hydrolysis of the carbamate ester, followed by reduction of the ketone group. The major metabolite is the 2-amino form, 2-amino-5-benzoylbenzimidazole, which, along with other hydroxy and hydroxyamino metabolites, is pharmacologically inactive and devoid of anthelmintic activity.[14] Because metabolism is the primary clearance mechanism, patients with impaired hepatic function or biliary obstruction may experience significantly higher plasma levels of the parent drug, necessitating caution and potential dose adjustments.[11]
The elimination of mebendazole is consistent with its poor absorption. The vast majority (over 90%) of an administered oral dose is excreted unchanged in the feces, having never been absorbed systemically.[1] The small, absorbed fraction is primarily cleared via the bile and subsequently excreted in the feces, with a minor portion (approximately 2-10%) eliminated in the urine, mostly as metabolites.[1] Some evidence suggests the drug may undergo a degree of enterohepatic recirculation.[13]
The plasma elimination half-life of mebendazole is relatively short, averaging 3 to 6 hours in individuals with normal hepatic function.[1] This half-life can be substantially prolonged in patients with cholestatic liver disease or other forms of severe hepatic impairment.[20]
Mebendazole is a cornerstone therapy for the treatment of intestinal worm infections, valued for its broad spectrum of activity against common nematodes and its generally favorable safety profile at standard doses. Its clinical use is well-established, with specific dosing regimens tailored to the type of parasitic infection.
Mebendazole is a broad-spectrum anthelmintic agent indicated for the treatment of single or mixed gastrointestinal infestations caused by a range of nematode species.[1] In the United States, the Food and Drug Administration (FDA) has approved its use in patients aged two years and older for the following infections [22]:
It is also used globally for other parasitic diseases, such as guinea worm infections and hydatid disease, although its poor systemic absorption makes other agents preferable for infections that extend beyond the gastrointestinal tract.[1] Off-label use for infections caused by tapeworms has also been reported.[27]
The efficacy of mebendazole varies depending on the target parasite. Clinical studies have demonstrated high parasitological cure rates for pinworm (95%), common roundworm (98%), and hookworm (96%). Its efficacy against whipworm is more moderate, with a reported cure rate of 68%.[23]
The dosing of mebendazole is highly dependent on the specific helminth being treated and the pharmaceutical formulation available (typically 100 mg or 500 mg chewable tablets).[30] The variability in these regimens is not arbitrary but reflects the drug's differential efficacy against various parasites. For easily treated luminal infections like pinworm, a simple, convenient single dose is sufficient. However, for more tenacious parasites like whipworm, a more intensive, multi-day regimen is required to achieve adequate cure rates, highlighting a clinical trade-off between adherence and maximal efficacy.
The introduction of a single 500 mg dose formulation aims to simplify treatment, particularly in the context of large-scale mass drug administration programs where adherence is a key determinant of success.[24] Standard dosing guidelines are summarized in Table 2.
Table 2: Dosing Regimens for Mebendazole in Approved Helminthic Infections
Indication (Parasite) | Patient Population | 100 mg Tablet Regimen | 500 mg Tablet Regimen | Clinical Notes |
---|---|---|---|---|
Pinworm (E. vermicularis) | Adults & Children ≥2 years | 100 mg as a single dose. | 500 mg as a single dose. | A second dose after 2-4 weeks is strongly recommended to prevent reinfection from hatched eggs. All household members should be treated simultaneously. 30 |
Roundworm (A. lumbricoides) | Adults & Children ≥2 years | 100 mg twice daily for 3 consecutive days. | 500 mg as a single dose. | If the patient is not cured 3 weeks after treatment, a second course may be necessary. 8 |
Whipworm (T. trichiura) | Adults & Children ≥2 years | 100 mg twice daily for 3 consecutive days. | 500 mg as a single dose. | Efficacy is moderate; a second course of treatment may be required if infection persists. 8 |
Hookworm (A. duodenale, N. americanus) | Adults & Children ≥2 years | 100 mg twice daily for 3 consecutive days. | 500 mg as a single dose. | If the patient is not cured 3 weeks after treatment, a second course may be necessary. 8 |
For pediatric patients, mebendazole is generally approved for use in children aged two years and older. However, it can be prescribed by a physician for children between 6 months and two years of age if the clinical benefit is deemed to outweigh the risks.[28] Dosing for children is typically the same as for adults.
Mebendazole is also used off-label at higher doses and for longer durations for more severe or systemic infections, such as capillariasis (200 mg twice daily for 20 days) and trichinellosis (200–400 mg three times daily for 3 days, followed by 400–500 mg three times daily for 10 days).[30]
The tablets are designed to be chewed but can also be swallowed whole or crushed and mixed with food to facilitate administration, especially in young children.[8] It can be taken with or without food, though absorption is enhanced with fatty meals.[32]
Successful treatment of helminthiasis requires more than just medication. Patient education on strict personal hygiene measures is critical to prevent reinfection and the transmission of parasites to others, a point of particular importance for the highly contagious pinworm.[23] Key recommendations include:
To effectively eradicate the infection within a household or community, it is standard practice to recommend simultaneous treatment for all family members or close contacts, regardless of whether they are symptomatic.[33]
Mebendazole is generally regarded as a safe and well-tolerated medication, particularly at the standard low doses used for treating common intestinal helminth infections. However, its safety profile shifts significantly with the administration of higher doses or for prolonged durations, revealing a dose-dependent pattern of toxicity that is critical to understand for its expanded use in systemic infections and oncology.
At recommended anthelmintic doses, adverse effects are typically mild, transient, and infrequent.[1]
The safety concerns associated with mebendazole escalate when it is administered at higher doses (e.g., for hydatid disease) or for extended periods, as is necessary for its investigational use in oncology. A clear dose-dependent shift in the primary dose-limiting toxicity is observed. While historical data from high-dose anthelmintic therapy pointed to bone marrow suppression as the most severe risk, recent clinical trials in oncology have identified hepatotoxicity as the more immediate concern at supra-pharmacological doses.
While mebendazole is a widely used and generally safe medication, there are specific circumstances where its use is contraindicated, along with important warnings and clinically significant drug interactions that healthcare professionals must consider to ensure patient safety.
The sole absolute contraindication for mebendazole is a history of known hypersensitivity (allergy) to mebendazole or any of the excipients present in the pharmaceutical formulation.[22]
Several important warnings and precautions are associated with the use of mebendazole:
Mebendazole is subject to several clinically significant drug-drug interactions, primarily related to its metabolism via the cytochrome P450 enzyme system. These are summarized in Table 3.
Table 3: Clinically Significant Drug Interactions with Mebendazole
Interacting Drug | Mechanism | Clinical Effect | Management Recommendation |
---|---|---|---|
Metronidazole | Pharmacodynamic Synergism (Mechanism not fully elucidated) | Increased risk of severe skin reactions: Stevens-Johnson Syndrome (SJS) and Toxic Epidermal Necrolysis (TEN). | Avoid concomitant use. 1 |
Cimetidine | Inhibition of Hepatic Metabolism (CYP450 enzymes) | Increases plasma concentrations of mebendazole, potentially increasing the risk of systemic toxicity. | Use with caution; monitor for adverse effects, especially with high-dose therapy. 8 |
Phenytoin, Carbamazepine | Induction of Hepatic Metabolism (CYP450 enzymes) | Decreases plasma concentrations of mebendazole, which may reduce its efficacy for systemic infections (e.g., hydatid disease) or in oncology. | Monitor for lack of efficacy. Dose adjustments may be necessary for systemic indications. 8 |
Deferiprone | Pharmacodynamic Synergism | Potential for additive risk of neutropenia or agranulocytosis. | Avoid concomitant use. If unavoidable, monitor absolute neutrophil count frequently. 8 |
Mebendazole and albendazole are the two primary benzimidazole anthelmintics used globally for the treatment of soil-transmitted helminth (STH) infections. While they share a similar mechanism of action and are often used interchangeably, head-to-head clinical trials have revealed crucial differences in their efficacy spectra. These differences have significant implications for both individual patient treatment and large-scale public health deworming strategies.
A key randomized controlled trial directly compared the efficacy of single-dose and triple-dose regimens of both mebendazole (500 mg) and albendazole (400 mg) against the major STHs.[47] The findings, summarized in Table 4, highlight a clear pattern of differential activity.
Table 4: Comparative Efficacy of Mebendazole vs. Albendazole for Major Soil-Transmitted Helminths
Parasite | Treatment Regimen | Mebendazole Cure Rate (95% CI) | Albendazole Cure Rate (95% CI) |
---|---|---|---|
Hookworm | Single Dose | 29% (20–45%) | 69% (55–81%) |
Hookworm | Triple Dose | 54% (46–71%) | 92% (81–98%) |
Whipworm | Single Dose | 40% | 34% |
Whipworm | Triple Dose | 71% (57–82%) | 56% |
Roundworm | Single Dose | >90% | >90% |
Data adapted from [47]
The comparative data strongly support the use of multi-day (triple-dose) regimens over the widely used single-dose approach to achieve high cure rates for both hookworm and whipworm infections.[47] The choice between a convenient single 500 mg dose and a more effective 3-day course of 100 mg twice daily represents a classic clinical trade-off between maximizing patient adherence and achieving the highest possible parasitological cure rate.
The nuanced differences in efficacy between mebendazole and albendazole are magnified at the population level and have profound implications for public health policy. The choice of drug for mass drug administration (MDA) programs should not be arbitrary but must be guided by local epidemiological data. Using an anthelmintic that is suboptimal for the predominant local parasite can lead to widespread treatment failure and the persistence of infection despite significant investment in deworming campaigns.
For example, exclusively using mebendazole in a region where hookworm is the primary endemic STH could result in a cure rate as low as 29% with a single-dose regimen, whereas using albendazole could achieve a 69% cure rate.[47] Scaled across a population of millions, this difference represents a massive disparity in public health impact. Therefore, an evidence-based approach is imperative:
In recent years, mebendazole has emerged as one of the most promising candidates for drug repurposing in oncology. A substantial body of preclinical evidence, coupled with early-phase clinical trials, suggests that this well-established anthelmintic possesses potent and multi-faceted anti-cancer properties.
Mebendazole embodies many of the ideal characteristics for a repurposed drug, making it an attractive and low-risk candidate for investigation in cancer therapy [13]:
Mebendazole has demonstrated significant anti-tumor activity in a wide array of preclinical models, including both in vitro cancer cell lines and in vivo animal xenograft models. Its efficacy has been shown against numerous cancer types, such as glioblastoma, medulloblastoma, colorectal cancer, breast cancer, lung cancer, pancreatic cancer, and adrenocortical carcinoma.[4]
Its anti-cancer effects are not mediated by a single mechanism but by a multi-pronged attack on several key pathways essential for tumor growth, survival, and metastasis [13]:
The promising preclinical data has led to the initiation of several clinical trials investigating mebendazole in cancer patients. The most mature data comes from a Phase 1 study in patients with brain tumors.
Table 5: Summary of Key Clinical Trials of Mebendazole in Oncology
Trial ID | Cancer Type | Phase | Regimen | Key Findings (Safety & Efficacy) |
---|---|---|---|---|
NCT01729260 | Newly Diagnosed High-Grade Glioma (Adult) | Phase 1 | Mebendazole (up to 200 mg/kg/day) + standard temozolomide. | Safety: Regimen was safe and well-tolerated long-term. Dose-limiting toxicity was reversible Grade 3 elevation of liver enzymes (ALT/AST). Efficacy: Median overall survival was 21 months. Median PFS was 13.1 months for patients on drug >1 month vs. 9.2 months for <1 month. Results are encouraging but not definitive. 26 |
NCI-2017-00550 | Recurrent/Refractory Pediatric Brain Tumors | Phase 1 | Mebendazole monotherapy dose escalation. | To determine the maximum tolerated dose (MTD) and safety profile in pediatric patients. 43 |
NCT03925662 | Colon Cancer | Phase 1/2 | Mebendazole in combination with other therapies. | Trial is underway to evaluate safety and efficacy. 26 |
In addition to formal trials, there are case reports of long-term disease control in patients with advanced adrenocortical carcinoma treated with mebendazole.[42] Some observational data also suggests that adding mebendazole to standard chemotherapy may slow disease progression in patients with metastatic colorectal cancer.[52]
The existing body of evidence strongly supports the continued investigation of mebendazole as a repurposed anti-cancer agent. Its ability to cross the blood-brain barrier makes it a particularly compelling candidate for both adult and pediatric brain cancers.[25] Its novel mechanism of HIF inhibition warrants further exploration as a strategy to overcome therapeutic resistance in a wide range of solid tumors.[49] Larger, randomized Phase 2 and Phase 3 clinical trials are now necessary to definitively establish its efficacy, determine optimal dosing and scheduling, and identify its precise role within the broader landscape of cancer therapy, whether as a monotherapy or in combination with existing treatments.
Mebendazole's journey from its development in the 1970s to its current global status reveals a bifurcated existence. It is simultaneously a low-cost, widely accessible staple of global public health and a niche, commercially fragile prescription product in some high-income countries. This unique regulatory and commercial history has significant implications for its future development, particularly for its potential repurposing in oncology.
Mebendazole was developed by Janssen Pharmaceutica in Belgium and first introduced for human use in 1971.[1] Its 100 mg chewable tablet formulation, under the brand name Vermox™, received its initial U.S. FDA approval in 1974 (NDA 017841).[24]
In a notable turn, marketing of the original Vermox™ was discontinued in the United States in 2006 for commercial reasons, not due to any safety or efficacy concerns.[24] The drug was later reintroduced to the U.S. market through new regulatory approvals, including Emverm™ (100 mg chewable tablet, approved in 2016) and a new formulation of Vermox™ Chewable (500 mg tablet, approved in 2016) under the 505(b)(2) pathway.[24] The 500 mg chewable tablet was also granted Orphan Drug Designation by the FDA for the treatment of soil-transmitted helminths.[24]
Mebendazole's status as a globally important medicine is solidified by its inclusion on the World Health Organization's List of Essential Medicines.[1] This status reflects its critical role in public health, particularly in low- and middle-income countries. Johnson & Johnson, the original developer, maintains a large-scale donation program, providing hundreds of millions of doses of 500 mg mebendazole tablets annually for mass deworming campaigns in endemic regions.[24]
The prescription status of mebendazole varies significantly around the world, reflecting different regulatory philosophies and public health needs:
This regulatory landscape highlights a significant disconnect. The drug's primary volume and public health impact are concentrated in developing nations, where it is an accessible commodity. In contrast, its presence in high-income markets like the U.S. is more tenuous and commercially driven. This lack of a strong commercial driver in major Western markets could pose a challenge to securing the substantial investment required for large-scale Phase 3 oncology trials, potentially leaving its development in this area reliant on academic and government-funded research.
Mebendazole is a synthetic compound produced through multi-step organic synthesis.[57] As a benzimidazole derivative, its synthesis involves the construction of the core heterocyclic ring system. A common industrial pathway utilizes 3,4-diaminobenzophenone as a key chemical intermediate.[11]
One described synthetic route begins with starting materials such as o-dichlorobenzene and benzoyl chloride. These undergo reactions of condensation and ammoniation to build the necessary precursors.[57] The final and characteristic step is a cyclization reaction. The 3,4-diaminobenzophenone intermediate is reacted with a reagent that provides the carbamate side chain, such as methyl cyanocarbamate or N-methoxycarbonyl-S-methylthiourea. This reaction closes the imidazole ring, forming the final mebendazole structure.[11] The synthesis and purification of key intermediates, such as o-chloraniline, are also critical steps in the overall manufacturing process.[58]
Published at: August 26, 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.