Small Molecule
C30H35F7N4O2
414910-27-3
Casopitant (DrugBank ID: DB06634) is an investigational small molecule drug developed by GlaxoSmithKline as a potent, selective, and centrally-acting neurokinin-1 (NK1) receptor antagonist.[1] Its primary therapeutic target was the prevention of chemotherapy-induced nausea and vomiting (CINV), a significant and debilitating side effect of cancer treatment. The development program for Casopitant progressed through extensive Phase III clinical trials, where it demonstrated robust and statistically significant efficacy. When added to the standard-of-care antiemetic regimen (a 5-HT3 receptor antagonist and a corticosteroid), both oral and intravenous formulations of Casopitant consistently improved the control of emesis in patients receiving both highly emetogenic chemotherapy (HEC) and moderately emetogenic chemotherapy (MEC).[3] The drug proved superior to control regimens, achieving a complete response (defined as no emesis and no use of rescue medication) in a significantly higher proportion of patients over the critical 5-day post-chemotherapy period.[3]
Despite this clear clinical efficacy, the trajectory of Casopitant was ultimately defined by its challenging pharmacokinetic and safety profile. The molecule is extensively metabolized, primarily by the cytochrome P450 3A4 (CYP3A4) enzyme system.[5] This heavy reliance on a single, highly variable metabolic pathway created a profound susceptibility to drug-drug interactions (DDIs). The pivotal safety concern emerged from a clinical study that revealed a significant prolongation of the QTc interval—a marker for increased risk of life-threatening cardiac arrhythmias—when Casopitant was co-administered with ketoconazole, a strong inhibitor of CYP3A4.[7] Given that the target patient population (individuals with cancer) is frequently polymedicated and often requires treatment with potent CYP3A4 inhibitors (e.g., azole antifungals), this DDI risk was deemed unmanageable.
Consequently, in September 2009, GlaxoSmithKline withdrew its marketing authorization application from the European Medicines Agency, citing the need for further safety assessments that would require considerable time to complete.[2] This decision effectively halted the global development of Casopitant for CINV. The drug was also briefly investigated for major depressive disorder, leveraging the role of the NK1 pathway in affective regulation, but this did not advance.[2] The history of Casopitant serves as a compelling case study in modern drug development, illustrating that potent pharmacodynamic activity and proven clinical efficacy can be completely negated by an unfavorable metabolic profile that introduces unacceptable safety risks in a vulnerable patient population. It underscores the principle that a drug's interaction with metabolic enzymes is as critical to its success as its interaction with its therapeutic target.
Casopitant is a synthetic, organic small molecule belonging to the phenylpiperidine class of compounds.[10] Its complex structure was designed to achieve high-affinity binding to the NK1 receptor and facilitate penetration of the blood-brain barrier. A comprehensive and unambiguous identification of the compound is established through its systematic nomenclature, standardized structural representations, and precise molecular formula.
The formal chemical name for Casopitant, according to the International Union of Pure and Applied Chemistry (IUPAC) nomenclature, is (2R,4S)-4-(4-acetylpiperazin-1-yl)-N-ethyl]-2-(4-fluoro-2-methylphenyl)-N-methylpiperidine-1-carboxamide.[1] This name precisely describes the stereochemistry and connectivity of all constituent atoms within the molecule. The drug has also been developed and studied as its mesylate salt form, casopitant mesilate or casopitant mesylate.[1]
For computational chemistry, database integration, and unambiguous structural referencing, several standardized identifiers are used to represent Casopitant:
These text-based representations allow for the precise digital encoding of the molecule's two-dimensional structure, including its specific stereoisomeric configuration.
The elemental composition of Casopitant is defined by its molecular formula, C30H35F7N4O2.[1] Based on this formula, the following molecular weights have been computed:
From a structural standpoint, Casopitant is classified as a phenylpiperidine. This core structure consists of a piperidine ring directly bonded to a phenyl group.[10] Its broader chemical taxonomy includes alternative parent classes such as trifluoromethylbenzenes, piperidinecarboxamides, N-alkylpiperazines, and aryl fluorides, reflecting the various functional groups appended to its central scaffold.[10]
The following table provides a consolidated reference for the key identifiers and physicochemical properties of Casopitant. This centralization of data from numerous chemical and pharmacological databases serves as a high-density information source for researchers and facilitates cross-referencing across different platforms.
Table 2.1: Comprehensive Identifiers and Properties of Casopitant
Property/Identifier Type | Value | Source(s) |
---|---|---|
Drug Name | Casopitant | 1 |
DrugBank ID | DB06634 | 1 |
CAS Number | 414910-27-3 | 1 |
IUPAC Name | (2R,4S)-4-(4-acetylpiperazin-1-yl)-N-ethyl]-2-(4-fluoro-2-methylphenyl)-N-methylpiperidine-1-carboxamide | 1 |
Synonyms | GW-679769, GW679769, Rezonic (proposed), Zunrisa (proposed) | 1 |
Molecular Formula | C30H35F7N4O2 | 1 |
Average Mass | 616.625 g/mol | 2 |
Monoisotopic Mass | 616.264823524 Da | 1 |
UNII | 3B03KPM27L | 1 |
ChEMBL ID | CHEMBL1672054 | 1 |
PubChem CID | 9917021 | 1 |
InChIKey | XGGTZCKQRWXCHW-WMTVXVAQSA-N | 1 |
Drug Type | Small Molecule | 1 |
Chemical Class | Phenylpiperidines | 10 |
The pharmacological activity of Casopitant is rooted in its highly specific interaction with the neurokinin-1 receptor system, a key pathway in the neurobiology of emesis. Its therapeutic effect is a direct consequence of its ability to competitively block the endogenous signaling molecule, Substance P, at critical sites within the central nervous system.
The emetic reflex, particularly the delayed phase of CINV, is heavily mediated by the tachykinin family of neuropeptides.[16] The most prominent member of this family is Substance P (SP), an undecapeptide that functions as a primary neurotransmitter and neuromodulator in both the central and peripheral nervous systems.[17] SP exerts its biological effects by binding to and activating its preferred receptor, the neurokinin-1 (NK1) receptor, also known as tachykinin receptor 1 (TACR1).[16] The NK1 receptor is a G protein-coupled receptor that, upon activation, initiates a phosphatidylinositol-calcium second messenger cascade.[10]
In the context of emesis, NK1 receptors are densely expressed in brain regions that form the central emetic pathway, including the nucleus tractus solitarius (NTS) and the area postrema (which contains the chemoreceptor trigger zone).[18] Cytotoxic chemotherapy agents can cause the release of SP from vagal afferent nerve endings in these brainstem centers.[16] The subsequent binding of SP to NK1 receptors triggers the downstream neuronal signaling that culminates in the complex physiological responses of nausea and vomiting.[16] While the acute phase of CINV (occurring within 24 hours of chemotherapy) is primarily driven by serotonin acting on 5-HT3 receptors, the delayed phase (occurring on days 2 through 5) is largely mediated by the SP/NK1 pathway.[17] This distinction provides the fundamental rationale for the development of NK1 receptor antagonists as a complementary and essential component of antiemetic therapy, specifically targeting the difficult-to-control delayed symptoms.
Casopitant's primary mechanism of action is as a potent, selective, and competitive antagonist of the human NK1 receptor.[1] By competitively binding to the NK1 receptor, Casopitant physically occupies the binding site, thereby preventing the endogenous ligand, Substance P, from activating it.[1] This blockade of the SP/NK1 interaction effectively interrupts the emetic signaling cascade at its source, resulting in a powerful antiemetic effect.[1] The high affinity of Casopitant for its target is demonstrated by bioactivity data, which report a binding affinity constant (
Ki) of 9.90 (-log[M]), indicating potent binding at nanomolar concentrations.[20] This high degree of selectivity is a crucial feature; unlike older antiemetics, Casopitant has little to no affinity for serotonin (5-HT3), dopamine, or corticosteroid receptors, meaning its therapeutic action is highly focused and it does not interfere with the mechanisms of other standard antiemetic agents.[18]
A defining characteristic of Casopitant and other successful small-molecule NK1 antagonists is their ability to exert their effects within the central nervous system. Early attempts to develop peptide-based NK1 antagonists were unsuccessful largely because these molecules were unable to cross the blood-brain barrier (BBB) and reach the critical emetic centers in the brainstem.[19] The molecular design of Casopitant successfully overcame this limitation. It is a brain-penetrant compound, a property essential for its antiemetic activity.[5] This ability to access central targets has been confirmed in both preclinical models and human studies using Positron Emission Tomography (PET).[16] These imaging studies visualize the drug's entry into the brain and confirm its occupancy of NK1 receptors at therapeutic doses, establishing a direct link between target engagement in the CNS and the observed clinical antiemetic response.[16] The successful engineering of a molecule with both high NK1 receptor affinity and the physicochemical properties needed for BBB penetration was the key pharmacological breakthrough that enabled the potent efficacy seen in clinical trials.
While the NK1 receptor is the primary therapeutic target of Casopitant, a secondary molecular interaction of profound clinical importance is with the enzyme Cytochrome P450 3A4 (CYP3A4).[5] Pharmacological profiling reveals a dual role for this interaction: Casopitant is both a
substrate for and a weak to moderate inhibitor of CYP3A4.[5] This means that the drug's own metabolic clearance is dependent on this enzyme, and it can simultaneously interfere with the metabolism of other drugs that are also cleared by CYP3A4. This characteristic is not merely a secondary feature but a central element of the drug's overall profile, creating a high intrinsic potential for clinically significant drug-drug interactions, which ultimately proved to be its critical liability.
Based on its mechanism and therapeutic use, Casopitant is classified under the following systems:
The disposition of Casopitant in the body has been extensively characterized through preclinical studies and, most definitively, a human clinical trial using radiolabeled [14C]casopitant.[22] This study provided a comprehensive assessment of the drug's absorption, distribution, metabolism, and excretion (ADME), revealing a profile characterized by rapid absorption, extensive tissue distribution, complex and nearly complete metabolism, and elimination primarily through the feces. This pharmacokinetic profile is fundamental to understanding both its clinical efficacy and the safety concerns that led to its discontinuation.
Following oral administration, Casopitant is rapidly absorbed.[19] A human study with radiolabeled drug demonstrated that the absorption of drug-related material from the gastrointestinal tract was nearly complete.[22] Preclinical data in ferrets indicated a time to maximum plasma concentration (
Tmax) of approximately 1 hour, and clinical observations in humans are consistent with rapid absorption.[19] The absolute oral bioavailability is substantial, reported to be in the range of 60% to 83%, indicating that despite some first-pass metabolism, a large fraction of the administered dose reaches systemic circulation.[21]
Once absorbed, Casopitant distributes extensively throughout the body. The volume of distribution (Vd) is reported to be 2.77 L/kg, a value that suggests significant partitioning from the plasma into peripheral tissues.[20] This is consistent with its lipophilic nature, which also facilitates its entry into the central nervous system.
Casopitant is highly bound to plasma proteins, with the unbound fraction (fu) estimated to be only 1%.[20] This high degree of protein binding means that only a small fraction of the drug in circulation is free to distribute into tissues and interact with its target receptors. However, its efficacy confirms that this small unbound fraction is sufficient to achieve therapeutic concentrations at the site of action.
As noted previously, a key feature of its distribution profile is its ability to effectively cross the blood-brain barrier.[21] Preclinical studies in ferrets using radiolabeled Casopitant confirmed that the parent compound was the predominant drug-related species found in brain tissue, accounting for approximately 76% of the radioactivity. Two major oxidative metabolites, M1 (hydroxylated) and M2 (ketone), were also detected in the brain, though at much lower levels (19% and 3%, respectively), indicating that the primary pharmacological activity in the CNS is attributable to the parent drug.[19]
Casopitant undergoes extensive and complex metabolism, with only negligible amounts of the parent drug being excreted unchanged.[22] The primary route of biotransformation is oxidation, a process mediated predominantly by the cytochrome P450 isoenzyme CYP3A4.[5] This heavy reliance on a single metabolic pathway is a critical feature of its pharmacokinetic profile.
The human radiolabel study identified two major circulating metabolites in plasma [22]:
Beyond these major circulating components, a complex pattern of additional metabolites was identified in plasma and excreta. The principal metabolic pathways include multiple oxidations at various positions on the molecule, loss of the N-acetyl group, N-demethylation, and modifications or cleavage of the piperazine ring structure.[22] A limited amount of Phase II metabolism also occurs, with some oxidized products undergoing conjugation with glucuronic acid, particularly those found in urine.[23]
The combination of nearly complete absorption, extensive first-pass metabolism, and primary clearance via a single enzyme (CYP3A4) creates a high-risk profile for drug-drug interactions. The drug's therapeutic effect is dependent on achieving and maintaining adequate plasma concentrations. However, these concentrations are highly susceptible to significant alteration by concomitant medications that either inhibit or induce CYP3A4 activity. Co-administration of a strong CYP3A4 inhibitor would be expected to block Casopitant's metabolism, leading to a sharp and potentially dangerous increase in its plasma levels. Conversely, co-administration of a strong CYP3A4 inducer would accelerate its clearance, potentially causing a loss of efficacy. This inherent vulnerability of its metabolic pathway proved to be a decisive clinical liability. Furthermore, the high intersubject variability (72%) observed in its oral clearance is a likely reflection of the well-known genetic and environmental variability of CYP3A4 expression and activity in the human population, adding another layer of unpredictability to its dosing.[19]
The elimination of Casopitant and its numerous metabolites occurs principally via the feces.[22] This was observed following both oral and intravenous administration of the radiolabeled drug, indicating that biliary excretion of metabolites into the gastrointestinal tract is the predominant route of clearance from the body.[23] Urinary excretion represents only a minor elimination pathway, accounting for less than 8% of the total administered radioactive dose.[22] Unchanged parent drug was never quantifiable in urine, further confirming the completeness of its metabolic clearance.[26]
Key pharmacokinetic parameters for Casopitant have been determined from clinical studies:
The clinical development program for Casopitant was extensive, culminating in multiple large-scale, multinational Phase III trials that rigorously evaluated its efficacy and safety. The primary focus of this program was the prevention of CINV, for which Casopitant was studied in both oral and intravenous formulations.[1] The drug was also explored in a Phase II study for a distinct neurological indication, major depressive disorder.
The core strategy of the CINV trials was to assess Casopitant as an additive therapy to the existing standard of care, which consisted of a 5-HT3 receptor antagonist (e.g., ondansetron) and a corticosteroid (dexamethasone).[3] This design aimed to demonstrate that a three-drug regimen including Casopitant was superior to the two-drug standard.
The efficacy of Casopitant in the most challenging setting—patients receiving HEC, defined as cisplatin-based regimens—was evaluated in a pivotal Phase III trial (NCT00431236).[4] This randomized, double-blind, placebo-controlled study enrolled 810 chemotherapy-naïve patients across 22 countries. Participants were randomized to receive the standard two-drug regimen (ondansetron and dexamethasone) plus one of the following: a placebo, a single 150 mg oral dose of casopitant on day 1, or a 3-day regimen of 90 mg intravenous casopitant on day 1 followed by 50 mg oral casopitant on days 2 and 3.[4]
The primary endpoint was the proportion of patients achieving a Complete Response (CR), stringently defined as no vomiting, no retching, and no use of rescue antiemetic medication during the entire 120-hour (5-day) period following chemotherapy initiation.[4] The results were definitive and demonstrated a strong therapeutic benefit for Casopitant.
This significant improvement in emetic control was sustained over multiple cycles of chemotherapy, confirming the durable efficacy of adding Casopitant to standard therapy for patients undergoing HEC.[4]
The benefit of Casopitant was also assessed in patients receiving MEC, a setting that still presents a significant burden of CINV. A large Phase III trial (NCT00366834) enrolled 1,840 patients, the majority of whom were women with breast cancer receiving anthracycline and cyclophosphamide (AC)-based regimens—a combination now considered by many guidelines to be highly emetogenic.[3] This trial had a four-arm design, comparing the standard two-drug control regimen against three different casopitant regimens: a single 150 mg oral dose on day 1; a 3-day oral regimen (150 mg on day 1, 50 mg on days 2-3); and a 3-day IV/oral regimen (90 mg IV on day 1, 50 mg on days 2-3).[3]
The primary endpoint was again the CR rate over the 120-hour overall period. The results mirrored the findings in the HEC setting, showing a consistent and statistically significant benefit across all casopitant arms.
A notable finding from this trial was the comparable efficacy of the convenient single-dose oral regimen to the more complex 3-day regimens. This suggested that a single administration of Casopitant could provide sustained protection against CINV for the full 5-day risk period in the MEC setting, a potential clinical and commercial advantage over the first-in-class NK1 antagonist, aprepitant, which requires a 3-day dosing schedule.[30] However, a consistent pattern observed across the CINV trials was that while Casopitant demonstrated profound efficacy in preventing the physical act of vomiting, it did not show a statistically significant reduction in the subjective patient-reported experience of nausea.[3] This suggests a potential divergence in the neurobiological control of these two distinct symptoms, with the SP/NK1 pathway being more critically involved in the motor act of emesis than in the sensation of nausea.
A further Phase III trial (NCT00601172) specifically investigated the efficacy of a single 90 mg intravenous dose of Casopitant in 710 patients receiving oxaliplatin-based MEC regimens.[32] The trial was designed to compare the efficacy of the three-drug regimen (IV casopitant, ondansetron, dexamethasone) against the standard two-drug control over a 120-hour period.[32] The results from this study, available in the clinical trial registry, provided additional evidence supporting the drug's efficacy in a specific subset of MEC patients.[33]
Table 5.1: Summary of Key Phase III CINV Trials for Casopitant
The clinical evidence supporting Casopitant's efficacy is extensive. The following table summarizes the design and primary outcomes of the pivotal trials, allowing for a rapid comparison of the consistent and robust benefit observed across different clinical settings.
Trial Identifier | Indication (Chemotherapy Type) | Patient Population (N) | Casopitant Arm(s) | Control Arm | Primary Endpoint | Primary Endpoint Result (CR % vs Control, p-value) | Source(s) |
---|---|---|---|---|---|---|---|
NCT00431236 | Highly Emetogenic (HEC) | 810 | 1) 150 mg PO single dose 2) 90 mg IV Day 1, 50 mg PO Days 2-3 | Placebo + Ondansetron + Dexamethasone | Complete Response (CR) in Overall Phase (0-120h) | 1) 86% vs 66% (p<0.0001) 2) 80% vs 66% (p=0.0004) | 4 |
NCT00366834 | Moderately Emetogenic (MEC) | 1840 | 1) 150 mg PO single dose 2) 150 mg PO Day 1, 50 mg PO Days 2-3 3) 90 mg IV Day 1, 50 mg PO Days 2-3 | Placebo + Ondansetron + Dexamethasone | Complete Response (CR) in Overall Phase (0-120h) | 1) 73% vs 59% (p<0.0001) 2) 73% vs 59% (p<0.0001) 3) 74% vs 59% (p<0.0001) | 3 |
NCT00601172 | Oxaliplatin-based MEC | 710 | 90 mg IV single dose | Placebo + Ondansetron + Dexamethasone | Complete Response (CR) in Overall Phase (0-120h) | Data available in registry, confirmed efficacy | 32 |
The role of Substance P and the NK1 receptor system extends beyond emesis to include the regulation of stress, anxiety, and mood.[21] This neurobiological link provided a strong rationale for investigating NK1 receptor antagonists as potential novel antidepressants.[2] In line with this hypothesis, Casopitant was evaluated for the treatment of Major Depressive Disorder in a Phase II clinical trial (NCT00413023).[9] While the completion of this trial indicates a serious exploration of this therapeutic avenue, the results have not been made publicly available, and this line of development was not pursued further, likely overshadowed by the subsequent discontinuation of the CINV program.[2]
While the clinical development program for Casopitant unequivocally established its efficacy, the ultimate fate of the drug was sealed by critical safety concerns stemming from its pharmacokinetic profile. The decision to halt its development was not a reflection of its performance against its therapeutic target but rather a proactive risk-management decision based on the potential for severe adverse events in its intended patient population.
Across the extensive Phase III program, which involved thousands of patients, the addition of Casopitant to standard antiemetic therapy was generally well-tolerated.[3] The safety profile was largely comparable between the casopitant-containing arms and the control arms receiving standard therapy alone. The most frequently reported adverse events were those commonly associated with the underlying malignancy and the administration of chemotherapy itself. These included fatigue, neutropenia, leukopenia, and anemia.[3] Importantly, the incidence of these events, as well as serious adverse events, was balanced across all treatment groups, suggesting that Casopitant did not add significant toxicity to the background of chemotherapy.[3]
The central safety issue that derailed Casopitant's path to approval was its interaction with the cytochrome P450 3A4 (CYP3A4) enzyme system. As established in the pharmacokinetic analysis, Casopitant is not only a substrate of CYP3A4 but also a weak to moderate inhibitor of the enzyme.[5] This created a high potential for clinically significant drug-drug interactions (DDIs).
The most alarming manifestation of this liability was identified in a dedicated clinical study designed to assess such interactions. The withdrawal assessment report from the European Medicines Agency (EMA) explicitly states that significant QTc prolongation was observed when Casopitant was co-administered with ketoconazole, a potent inhibitor of CYP3A4.[7] QTc prolongation is a well-established surrogate marker for an increased risk of Torsades de Pointes, a rare but potentially fatal cardiac arrhythmia. This finding represented a critical safety signal, demonstrating that inhibiting the primary metabolic pathway of Casopitant could lead to a dangerous accumulation of the drug and a direct, adverse effect on cardiac repolarization.
This single finding had profound implications for the drug's viability. The target patient population—individuals undergoing cancer chemotherapy—is characterized by frequent polypharmacy and a high likelihood of receiving concomitant medications that are strong CYP3A4 inhibitors. For example, azole antifungal agents (like ketoconazole, itraconazole, or posaconazole) are commonly used to prevent or treat fungal infections in patients with neutropenia, a frequent complication of chemotherapy. The risk of inadvertently triggering a life-threatening cardiac event by co-administering Casopitant with one of these common supportive care medications was deemed unacceptably high. Consequently, the clinical recommendation was that the administration of strong CYP3A4 inhibitors should be avoided in patients treated with Casopitant.[7] However, in real-world clinical practice, managing such a restriction in a complex patient population would be extremely challenging and fraught with risk.
On the strength of its positive Phase III efficacy data, GlaxoSmithKline (GSK) filed a marketing authorisation application with the EMA in July 2008, seeking approval for Casopitant under the proposed trade name Zunrisa.[2] A New Drug Application was also filed in the United States under the proposed trade name Rezonic.[34]
However, as the review process progressed and the full safety data, including the critical DDI study, were evaluated, the severity of the QTc prolongation risk became clear. In September 2009, GSK formally notified the EMA of its decision to withdraw the application for Zunrisa.[2] The company's official statement explained that the withdrawal was based on an internal assessment that "further safety data would be required to support the registration of casopitant on a worldwide basis and that it would take considerable time to produce these data".[8] This regulatory language signaled the discovery of a major safety impediment that could not be readily mitigated without substantial additional, and likely unfeasible, clinical investigation. All ongoing global applications were subsequently withdrawn, and the development of Casopitant for CINV was discontinued.[8]
To fully appreciate the factors that determined the fate of Casopitant, it is essential to compare its profile with that of other key drugs in the NK1 receptor antagonist class, particularly the approved agents Aprepitant and Rolapitant. This comparative analysis reveals that while all three drugs effectively engage the same therapeutic target, subtle but critical differences in their pharmacokinetic and metabolic properties led to divergent clinical and regulatory outcomes. The success or failure in this class was ultimately dictated less by efficacy and more by the nature and manageability of each drug's DDI profile.
Aprepitant was the first NK1 receptor antagonist to receive regulatory approval, establishing the clinical utility of this drug class.[19]
Rolapitant was developed later and represents an attempt to improve upon the pharmacokinetic profile of earlier agents.
The development pathways of these three drugs illustrate a clear evolution in the design philosophy for this class. Efficacy at the NK1 receptor was the initial bar. The next challenge was optimizing the pharmacokinetic and DDI profile. Aprepitant demonstrated that a complex but manageable DDI profile was acceptable. Rolapitant's design shows a deliberate effort to engineer a "cleaner" profile with respect to CYP3A4, the most common drug-metabolizing enzyme. Casopitant, despite its efficacy, possessed a DDI profile that fell into an unacceptable risk category. Its specific interaction with CYP3A4 inhibitors was directly linked to a severe cardiac safety signal that could not be easily mitigated in the intended patient population, ultimately making its risk-benefit calculus unfavorable.
Table 7.1: Comparative Profile of Key NK1 Receptor Antagonists
The following table provides a side-by-side comparison of the key features of Aprepitant, Rolapitant, and Casopitant. This format starkly highlights the critical differences in pharmacokinetics and drug interactions that determined their divergent regulatory and clinical fates.
Feature | Aprepitant | Rolapitant | Casopitant |
---|---|---|---|
Regulatory Status | Approved | Approved | Development Discontinued |
Half-life (t1/2) | 9–13 hours | ~180 hours | ~16 hours |
CINV Dosing Regimen | 3-day (Oral) | Single Dose (Oral) | Single or 3-day (Investigational) |
Primary Metabolism | CYP3A4 (major), CYP1A2, CYP2C19 | CYP3A4 | CYP3A4 |
CYP3A4 Interaction | Substrate, Moderate Inhibitor, Inducer | Substrate Only | Substrate, Weak-to-Moderate Inhibitor |
Other Key CYP Interaction | Inducer of CYP2C9 | Moderate Inhibitor of CYP2D6 | None well-defined, but QTc risk with CYP3A4 inhibition |
Key DDI Consequence | Requires dexamethasone dose adjustment; many interactions | Contraindicated with thioridazine, pimozide (CYP2D6 substrates) | Significant QTc prolongation with strong CYP3A4 inhibitors |
The clinical development and subsequent discontinuation of Casopitant represent a significant and instructive chapter in the history of supportive care in oncology. The extensive data accumulated throughout its lifecycle paint a clear picture of a molecule with undeniable pharmacodynamic potency and robust clinical efficacy, which was ultimately undone by an intractable pharmacokinetic liability.
The Phase III clinical program for Casopitant provided unequivocal evidence of its superiority over the standard antiemetic regimens of the time. In both highly and moderately emetogenic chemotherapy settings, the addition of Casopitant led to a clinically meaningful and statistically significant increase in the number of patients protected from vomiting and the need for rescue medication. Its efficacy, particularly with a convenient single-dose oral regimen in the MEC trial, positioned it as a potentially valuable new tool for managing one of the most feared side effects of cancer treatment.
However, the failure of Casopitant was not a failure of efficacy but a failure of safety rooted in its metabolism. The drug's heavy dependence on CYP3A4 for clearance, combined with its own inhibitory effect on this enzyme, created a precarious situation. The discovery that this interaction could lead to significant and dangerous QTc interval prolongation when combined with common CYP3A4 inhibitors was the decisive blow. For a drug intended for a vulnerable, polymedicated cancer patient population, the risk of inducing a fatal cardiac arrhythmia was a liability that could not be justified by its antiemetic benefit, however substantial.
The story of Casopitant offers several critical lessons for pharmaceutical development. Firstly, it underscores that in a therapeutic area with existing effective treatments, a new agent must not only demonstrate efficacy but also offer a comparable or superior safety profile. Casopitant, while highly effective, introduced a new and severe safety risk that was not as prominent with the first-in-class agent, aprepitant. Secondly, it highlights the paramount importance of a thorough and early characterization of a drug's metabolic pathways and DDI potential. The "cleanliness" of a drug's pharmacokinetic profile—its freedom from reliance on a single, easily modulated enzyme and its lack of potent inhibitory or inducing effects—is often the ultimate determinant of its success. The contrast with Rolapitant, which was designed to avoid CYP3A4 inhibition, is particularly telling.
While there was brief exploration of Casopitant for major depressive disorder, its development for any indication is now highly improbable. The shadow of its cardiac safety risk would necessitate an extremely high bar for any future investigation. Therefore, the primary legacy of Casopitant will be as a powerful cautionary tale in drug development and as a rich source of clinical and pharmacological data. It remains a textbook example of how a molecule's journey through the human body is as important as its ability to reach its target, and how a flaw in the former can render the latter irrelevant.
Published at: August 29, 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.