C19H24N2O
135729-56-5
Nausea caused by Chemotherapy, Post Operative Nausea and Vomiting (PONV), Acute chemotherapy-induced nausea and vomiting, Delayed chemotherapy-induced nausea and vomiting
Palonosetron is a second-generation serotonin 5-HT3 receptor antagonist distinguished by a unique pharmacological profile and superior clinical efficacy in the prevention of nausea and vomiting, particularly those induced by chemotherapy (CINV) and surgery (PONV). Chemically defined as the pure (S,S)-stereoisomer of a complex isoquinoline derivative, its structure confers a significantly higher binding affinity for the 5-HT3 receptor and facilitates unique allosteric interactions, which are not observed with first-generation agents like ondansetron or granisetron. These pharmacodynamic properties, combined with its exceptionally long pharmacokinetic half-life of approximately 40 hours, provide the basis for its sustained antiemetic activity.
Clinically, Palonosetron is the only 5-HT3 receptor antagonist monotherapy approved by the U.S. Food and Drug Administration (FDA) for the prevention of delayed CINV associated with moderately emetogenic chemotherapy (MEC). Pivotal clinical trials have consistently demonstrated its superiority over first-generation antagonists in providing a complete response—defined as no emesis and no rescue medication—over a 120-hour period. Its efficacy is well-established for acute CINV following both MEC and highly emetogenic chemotherapy (HEC), for PONV prevention up to 24 hours, and in pediatric populations as young as one month of age. Palonosetron serves as a foundational component in modern antiemetic regimens, where its efficacy is enhanced by co-administration with corticosteroids and neurokinin-1 (NK-1) receptor antagonists. This has led to its inclusion in guideline-recommended triple-therapy regimens and its development as part of a fixed-dose combination product, Akynzeo (palonosetron/netupitant).
The safety profile of Palonosetron is well-characterized and favorable. The most common adverse events are mild-to-moderate headache and constipation. Critically, it exhibits a superior cardiac safety profile compared to older agents, with a clinically insignificant effect on the QTc interval at therapeutic doses, a key advantage in polymedicated oncology patients. Warnings and precautions are in line with the drug class and include the potential for hypersensitivity reactions and Serotonin Syndrome, particularly when co-administered with other serotonergic medications. Its single-dose administration for each indication simplifies treatment protocols and improves patient convenience. Developed by Helsinn Healthcare SA and marketed under brand names including Aloxi®, Palonosetron's successful regulatory journey and strategic lifecycle management have established it as a standard of care and a cornerstone of modern antiemetic therapy.
The precise and unambiguous identification of a pharmaceutical agent is fundamental to its study and clinical application. This section provides a definitive chemical and physical profile of Palonosetron, detailing its nomenclature, structural characteristics, and key identifiers used across scientific and regulatory domains.
Palonosetron is known by a variety of chemical names, synonyms, and database identifiers that ensure its consistent recognition in global literature and regulatory filings.
The therapeutic activity of Palonosetron is a direct consequence of its specific three-dimensional structure and its resulting physical and chemical properties.
The chemical synthesis of Palonosetron is a multi-step process designed to produce the specific (S,S)-stereoisomer required for clinical activity. The manufacturing process must be stereoselective to avoid contamination with other, less active or potentially harmful isomers.
The following table provides a consolidated summary of the key chemical and physical identifiers for Palonosetron.
Table 2.1: Chemical and Physical Identifiers for Palonosetron
Property | Value | Source(s) |
---|---|---|
IUPAC Name | (3aS)-2-octan-3-yl]-2,3,3a,4,5,6-hexahydro-1H-benzo[de]isoquinolin-1-one | 1 |
DrugBank ID | DB00377 | 1 |
CAS Number | 135729-56-5 (free base) | 4 |
Molecular Formula | C19H24N2O | 2 |
Molecular Weight | 296.41 g/mol | 2 |
Stereochemistry | (S,S)-stereoisomer | 2 |
Physical Form | Off-white powder | 5 |
Melting Point | 87 - 88 °C | 2 |
InChIKey | CPZBLNMUGSZIPR-NVXWUHKLSA-N | 2 |
Oral Bioavailability | 97% | 2 |
The clinical utility and superiority of Palonosetron are rooted in its distinct pharmacological properties. As a second-generation 5-HT3 receptor antagonist, it possesses unique pharmacodynamic and pharmacokinetic characteristics that differentiate it from older agents in its class. This section dissects its mechanism of action at the molecular level and describes its journey through the body.
The pharmacodynamics of Palonosetron explain how it interacts with its biological target to produce its therapeutic antiemetic effect. Its mechanism is defined by high selectivity, potent affinity, and a unique mode of receptor interaction.
The fundamental mechanism of Palonosetron is the selective blockade of the serotonin 5-HT3 receptor, a ligand-gated ion channel.[1] Emesis, particularly CINV, is initiated in large part by the release of serotonin from enterochromaffin cells in the gastrointestinal (GI) tract following damage from cytotoxic chemotherapy.[1] This released serotonin activates 5-HT3 receptors located on peripheral vagal afferent nerves, which transmit signals to the medullary vomiting center in the brainstem. Serotonin also acts centrally on 5-HT3 receptors in the chemoreceptor trigger zone (CTZ) of the area postrema, a region that detects emetogenic substances in the blood.[8]
Palonosetron exerts its antiemetic activity by competitively inhibiting the binding of serotonin to these 5-HT3 receptors at both peripheral and central sites.[1] By blocking these receptors, it interrupts the signaling cascade that leads to the sensation of nausea and the vomiting reflex.[8] A key feature of Palonosetron is its high specificity; it has little to no affinity for other neurotransmitter receptors, such as other serotonin receptor subtypes (5-HT1, 5-HT2, 5-HT4), dopamine, adrenergic, histamine, or opioid receptors.[1] This high selectivity is crucial for its favorable safety profile, as it avoids off-target effects that could lead to unwanted side effects like sedation or extrapyramidal symptoms.
What elevates Palonosetron to a second-generation agent is not just its primary mechanism but the unique nature of its interaction with the 5-HT3 receptor. This interaction is characterized by two key features that contribute to its enhanced and prolonged efficacy.
First, Palonosetron exhibits a remarkably high binding affinity for the 5-HT3 receptor. This affinity is reported to be at least 30-fold greater than that of first-generation antagonists like ondansetron and granisetron.[9] This means that Palonosetron binds more tightly and effectively to the receptor, allowing for potent blockade at lower concentrations and contributing to its superior clinical efficacy observed in head-to-head trials.[8]
Second, and perhaps more significantly, Palonosetron demonstrates a unique molecular interaction with the receptor. Evidence suggests that it engages in allosteric interactions and exhibits positive cooperativity.[13] Unlike a simple competitive antagonist that just physically blocks the binding site, an allosteric modulator binds to a different site on the receptor and induces a conformational change. This change can alter the receptor's function, in this case, locking it in a prolonged, non-functional state that is resistant to activation by serotonin. This allosteric mechanism, which is not observed with first-generation agents, is thought to be a primary contributor to the drug's long duration of action, allowing it to suppress the emetic reflex long after peak plasma concentrations have passed.[13] This sophisticated binding mechanism provides a clear pharmacodynamic explanation for why Palonosetron is uniquely effective against delayed CINV.
The pharmacokinetic profile of Palonosetron describes its absorption, distribution, metabolism, and excretion. Its most notable feature is an exceptionally long elimination half-life, which works in concert with its potent pharmacodynamics to provide sustained antiemetic coverage from a single dose.
Palonosetron is formulated for both intravenous and oral administration.[2] When taken orally, it is well absorbed from the gut and exhibits excellent bioavailability, with studies showing that 97% of the oral dose reaches systemic circulation.[2] Following oral administration, peak plasma concentrations (
Cmax) are typically achieved in approximately 5.1 hours (Tmax), and this absorption is not affected by the presence of food.[2]
Once in the bloodstream, Palonosetron distributes extensively throughout the body. This is evidenced by its large apparent volume of distribution (Vd), which is approximately 8.3 L/kg.[1] This large value indicates that the drug does not remain confined to the bloodstream but partitions widely into body tissues. Palonosetron exhibits moderate binding to plasma proteins, with approximately 62% of the drug being bound.[1] The remaining unbound fraction is free to distribute to target tissues and interact with 5-HT3 receptors.
Approximately half (50%) of an administered dose of Palonosetron is metabolized, primarily in the liver.[1] This metabolism is mediated by the cytochrome P450 (CYP) enzyme system. The primary isoenzyme involved is CYP2D6, with minor contributions from CYP3A4 and CYP1A2.[1] This involvement of multiple CYP pathways may contribute to a lower risk of clinically significant drug-drug interactions, as the inhibition of a single pathway would not completely block the drug's metabolism.
The metabolic process yields two main metabolites: an N-oxide derivative and a 6S-hydroxy derivative.[2] Crucially, both of these metabolites are pharmacologically insignificant, possessing less than 1% of the 5-HT3 receptor antagonist activity of the parent compound, Palonosetron.[2] This means that the therapeutic effect is almost entirely attributable to the unchanged drug.
Palonosetron and its inactive metabolites are eliminated from the body primarily through the kidneys.[2] Following a single intravenous dose, about 80% of the dose is recovered in the urine over 144 hours, with approximately 40% of this being the unchanged parent drug.[2] A smaller fraction is eliminated via the feces.[2]
The defining pharmacokinetic feature of Palonosetron is its exceptionally long terminal elimination half-life (t1/2). In healthy individuals, the half-life is approximately 37 hours, and in cancer patients, it is even longer, averaging around 40 to 48 hours.[2] In some patients, the half-life can exceed 100 hours.[2] This is in stark contrast to first-generation 5-HT3 antagonists, whose half-lives are typically in the range of 2 to 15 hours.[2] This prolonged half-life ensures that therapeutically effective plasma concentrations of Palonosetron are maintained for several days following a single administration, providing the sustained coverage necessary to prevent both acute (0-24 hours) and delayed (24-120 hours) CINV.
The synergy between the drug's pharmacodynamics and pharmacokinetics is the core of its clinical success. The long half-life (a pharmacokinetic property) ensures the drug is present at the receptor for an extended period, while the high-affinity, allosteric binding (a pharmacodynamic property) ensures that its inhibitory effect on the receptor is profound and persistent. Together, these attributes provide a robust and prolonged antiemetic effect that is superior to that of older agents.
The following table compares the key pharmacological properties of Palonosetron with those of representative first-generation 5-HT3 antagonists.
Table 3.1: Comparative Pharmacological Profile of 5-HT3 Receptor Antagonists
Feature | Palonosetron | Ondansetron | Granisetron |
---|---|---|---|
Generation | Second | First | First |
Receptor Binding Affinity | Very High (>30x higher than 1st gen) | High | High |
Allosteric Interaction | Yes (Positive Cooperativity) | No | No |
Elimination Half-Life (t1/2) | ~40 hours | ~4-6 hours | ~9 hours |
Primary Metabolic Pathway | CYP2D6 (major), CYP3A4, CYP1A2 | CYP3A4, CYP1A2, CYP2D6 | CYP3A4 |
Key Differentiator | Prolonged half-life and unique allosteric binding leading to superior efficacy in delayed CINV | Standard for acute CINV, less effective for delayed CINV | Standard for acute CINV, less effective for delayed CINV |
Source(s) | 2 | 9 | 9 |
The clinical value of Palonosetron is defined by its proven efficacy in preventing nausea and vomiting across a range of challenging clinical scenarios. Its development and subsequent clinical trial program established a new benchmark for antiemetic therapy, particularly by demonstrating robust control over the delayed phase of CINV. This section details its approved indications and analyzes the pivotal clinical trial data that underpin its use.
Palonosetron is approved by major regulatory bodies, including the U.S. FDA and the European Medicines Agency (EMA), for several key indications in both adult and pediatric populations.
Palonosetron is a cornerstone therapy for the prevention of CINV, with specific approvals based on the emetogenic potential of the chemotherapy regimen:
Palonosetron is indicated for the prevention of PONV for up to 24 hours following surgery.[1] Its use is recommended in situations where PONV must be avoided, even if the baseline risk is low.[18] Efficacy beyond the initial 24-hour postoperative period has not been formally established.[16]
The use of Palonosetron has been extended to younger patients based on dedicated clinical studies. It is approved for children aged 1 month to less than 17 years for the prevention of acute nausea and vomiting associated with initial and repeat courses of emetogenic cancer chemotherapy, including HEC.[16] This approval provides a vital therapeutic option for one of the most vulnerable patient populations.
The approvals and guideline recommendations for Palonosetron are built on a foundation of rigorous, large-scale, randomized controlled trials. These studies not only established its efficacy but also demonstrated its superiority over the previous standard of care.
Two landmark Phase III, double-blind trials definitively established Palonosetron's superiority in the MEC setting.
These trials were groundbreaking because they were the first to show a clinically and statistically significant advantage of one 5-HT3 antagonist over others, particularly in the difficult-to-treat delayed phase.
In the HEC setting, where the risk of emesis is extremely high, a pivotal Phase III trial compared a single dose of Palonosetron (0.25 mg) with ondansetron (32 mg) in 667 patients, the majority of whom also received prophylactic dexamethasone.[15] The study found that Palonosetron was non-inferior to ondansetron in preventing acute CINV.[20] More importantly, in the subgroup of patients who received concomitant dexamethasone—which reflects modern clinical practice—Palonosetron demonstrated superior efficacy over ondansetron in controlling emesis throughout the entire 5-day post-chemotherapy period, with statistically significant advantages in the delayed and overall phases.[9]
To provide a more robust estimate of its relative efficacy, a pooled analysis of patient-level data from four large Phase III trials was conducted.[21] This analysis included 1,787 patients treated with Palonosetron and 1,175 treated with an older 5-HT3 antagonist (ondansetron, dolasetron, or granisetron). The results overwhelmingly confirmed the findings of the individual trials. CR rates were significantly higher for Palonosetron in the delayed period (57% vs. 45%,
p<0.0001) and the overall 0-120 hour period (51% vs. 40%, p<0.0001).[21] This large-scale analysis solidified Palonosetron's position as the most effective single-agent 5-HT3 antagonist for comprehensive CINV prevention.
Modern CINV prevention relies on multi-modal therapy targeting different emetic pathways. Palonosetron is a preferred backbone for these regimens.
The approval for PONV was based on Phase III trials comparing Palonosetron to placebo. In a pivotal study involving 574 patients undergoing gynecologic or abdominal laparoscopic surgery, a single 0.075 mg intravenous dose of Palonosetron given before anesthesia resulted in a significantly higher CR rate (no emesis, no rescue medication) in the first 24 hours compared to placebo (42.8% vs. 25.9%, p=0.0035).[15]
The pediatric approval was supported by a study that demonstrated a single 20 mcg/kg dose of Palonosetron was as effective as a standard ondansetron-based regimen in preventing acute CINV in patients from 1 month to less than 17 years of age.[19] The CR rate in the first 24 hours was 59.4% for the Palonosetron group, meeting the study's primary endpoint of non-inferiority.[19]
The clinical trial program for Palonosetron was instrumental in shifting the paradigm of antiemetic research. By focusing on endpoints that captured the full 5-day period of risk, particularly the delayed phase, the studies established a new and higher standard for efficacy. This changed clinical expectations from simply controlling acute vomiting to maintaining a vomit-free and low-nausea state for patients throughout their most vulnerable period, significantly improving quality of life.[15]
The following table summarizes the key results from the pivotal Phase III trials that established the efficacy of Palonosetron in CINV.
Table 4.1: Summary of Pivotal Phase III Clinical Trials for Palonosetron in CINV
Trial Reference / Focus | Patient Population (Emetogenicity) | N | Intervention | Comparator | Primary Endpoint | Key Result (Complete Response: Palonosetron vs. Comparator) | Source(s) |
---|---|---|---|---|---|---|---|
Gralla et al. 2003 | MEC | 570 | Palonosetron 0.25 mg IV | Ondansetron 32 mg IV | CR in Acute Phase | Acute (0-24h): 81.0% vs. 68.6% (p=0.008) Delayed (>24-120h): 74.1% vs. 55.1% (p<0.001) | 15 |
Eisenberg et al. 2003 | MEC | 592 | Palonosetron 0.25 mg IV | Dolasetron 100 mg IV | CR in Acute Phase | Acute (0-24h): 63.0% vs. 52.9% (p=0.049) Delayed (>24-120h): 54.0% vs. 38.7% (p=0.004) | 15 |
Aapro et al. 2006 | HEC (Cisplatin) | 667 | Palonosetron 0.25 mg IV | Ondansetron 32 mg IV | CR in Acute Phase | Acute (0-24h): 59.2% vs. 57.0% (Non-inferior) Delayed (>24-120h with Dexamethasone): 42.0% vs. 28.6% (p=0.021) | 9 |
Boccia et al. 2013 (Pooled) | MEC/HEC | 2,962 | Palonosetron 0.25/0.75 mg IV | Ondansetron/Dolasetron/Granisetron | CR in Acute, Delayed, Overall | Delayed (>24-120h): 57% vs. 45% (p<0.0001) Overall (0-120h): 51% vs. 40% (p<0.0001) | 21 |
A thorough understanding of a drug's safety profile is paramount to its appropriate clinical use. Palonosetron is generally well-tolerated, with a safety profile that is comparable or superior to older agents in its class. This section provides a comprehensive overview of its adverse effects, critical warnings and precautions, and clinically significant drug interactions.
The adverse reactions associated with Palonosetron have been characterized through extensive clinical trials in both adult and pediatric populations for CINV and PONV indications.
The product labeling for Palonosetron includes several critical warnings and precautions to guide its safe use and mitigate potential risks.
The potential for drug-drug interactions with Palonosetron is relatively low, which is a favorable characteristic for its use in polymedicated cancer patients.
The practical application of Palonosetron in the clinical setting is guided by its available formulations and specific, evidence-based dosing regimens for each indication and patient population. The drug's long half-life is a key determinant of its simple, single-dose administration schedule, which represents a significant advantage in convenience and compliance.
Palonosetron is commercially available in several ready-to-use formats, eliminating the need for complex reconstitution or dilution.
The dosing of Palonosetron is indication-specific and differs between adult and pediatric populations. The single-dose regimen for each therapeutic event (a cycle of chemotherapy or a surgical procedure) is a direct clinical benefit of its prolonged pharmacokinetic profile.
To ensure safety and efficacy, specific administration procedures must be followed:
The following table provides a consolidated clinical reference for the recommended dosing and administration of intravenous Palonosetron.
Table 6.1: Recommended Dosing and Administration for Intravenous Palonosetron
Indication | Patient Population | Dose | Route | Administration Details | Source(s) |
---|---|---|---|---|---|
CINV Prevention | Adults | 0.25 mg | IV Infusion | Infuse over 30 seconds, ~30 minutes before chemotherapy. | 17 |
CINV Prevention | Pediatrics (1 month to <17 years) | 20 mcg/kg (Max 1.5 mg) | IV Infusion | Infuse over 15 minutes, ~30 minutes before chemotherapy. | 17 |
PONV Prevention | Adults | 0.075 mg | IV Injection | Inject over 10 seconds, immediately before induction of anesthesia. | 17 |
The commercial success and widespread clinical adoption of Palonosetron are the result of a robust clinical development program and a strategic approach to lifecycle management. This section details the drug's key brand names, manufacturers, and its regulatory history, which illustrates its evolution from a novel standalone agent to a foundational component of next-generation antiemetic therapies.
Palonosetron is marketed globally under several brand names, both as a single agent and as part of a combination product.
The regulatory timeline of Palonosetron highlights a well-executed strategy of initial approval followed by indication expansions and formulation innovations to maximize its clinical utility and commercial longevity.
This regulatory pathway demonstrates a highly successful lifecycle management strategy. The initial approval established Palonosetron's superiority. Subsequent expansions into new indications (PONV), populations (pediatrics), and formulations (oral) broadened its clinical reach. The development of the Akynzeo® franchise was a pivotal strategic move. By leveraging Palonosetron's best-in-class profile as the backbone for a new, patented combination product, Helsinn effectively extended the commercial life of the franchise, addressed evolving clinical guidelines, and provided a more convenient therapeutic option for patients and providers.
The following table chronologically summarizes the key regulatory milestones in the history of Palonosetron and its related combination products.
Table 7.1: Key Regulatory Milestones for Palonosetron and Related Products
Date | Regulatory Body | Action | Product | Details of Action | Source(s) |
---|---|---|---|---|---|
July 25, 2003 | U.S. FDA | Initial Approval | Aloxi® Injection | Prevention of CINV in adults. | 39 |
Feb 29, 2008 | U.S. FDA | Indication Expansion | Aloxi® Injection | Prevention of PONV in adults. | 25 |
Aug 22, 2008 | U.S. FDA | New Formulation Approval | Aloxi® Capsules | Oral formulation for CINV prevention. | 29 |
May 28, 2014 | U.S. FDA | Population Expansion | Aloxi® Injection | Prevention of acute CINV in pediatric patients (≥1 month old). | 19 |
Oct 10, 2014 | U.S. FDA | New Product Approval | Akynzeo® Capsules | Fixed-dose oral combination of netupitant/palonosetron for CINV. | 24 |
May 26, 2016 | EMA | Initial Authorization | Palonosetron Accord | First marketing authorisation for a generic version in the EU. | 14 |
Apr 19, 2018 | U.S. FDA | New Product Approval | Akynzeo® for Injection | Fixed-dose IV combination of fosnetupitant/palonosetron for CINV. | 31 |
Palonosetron represents a significant advancement in antiemetic pharmacotherapy and a paradigm of successful rational drug design. Its development and clinical validation have fundamentally altered the standards and expectations for managing chemotherapy-induced and postoperative nausea and vomiting. The comprehensive analysis of its chemical, pharmacological, clinical, and regulatory profile reveals a molecule whose superiority is not incidental but is deeply rooted in its unique scientific attributes.
The journey from its chemical structure to its clinical performance is a clear and logical progression. The specific (S,S)-stereochemistry of its complex isoquinoline backbone was engineered to optimize its interaction with the 5-HT3 receptor. This resulted in a pharmacodynamic profile characterized by exceptionally high binding affinity and a unique allosteric binding mechanism, features that confer a more profound and durable receptor blockade than that of its predecessors. This potent molecular action is complemented perfectly by its distinctive pharmacokinetic profile, most notably an extended elimination half-life of approximately 40 hours. This synergy—a drug that binds more tightly and for longer at its target, while also remaining in the body for a prolonged period—is the definitive explanation for its superior clinical efficacy.
The clinical trial evidence is unequivocal. Palonosetron was the first 5-HT3 antagonist to demonstrate statistically significant superiority over first-generation agents in preventing CINV, particularly in the challenging delayed phase. This achievement redefined the goals of antiemetic therapy, shifting the focus from merely controlling acute emesis to providing comprehensive protection over the entire 5-day period of risk, thereby substantially improving patient quality of life. Its well-documented efficacy, combined with a favorable safety profile highlighted by a lack of clinically significant cardiac effects at therapeutic doses, has established it as a preferred agent in numerous clinical guidelines.
Ultimately, Palonosetron is more than just another antiemetic. It is a foundational agent in modern supportive cancer care. Its successful lifecycle management, culminating in its role as the cornerstone of the fixed-dose combination product Akynzeo, showcases its evolution from a standalone drug to an integral component of guideline-directed, multi-modal antiemetic strategies. In conclusion, Palonosetron stands as a testament to how targeted improvements in pharmacology can translate directly into meaningful and superior clinical outcomes, solidifying its position as a standard of care for preventing some of the most distressing side effects of cancer treatment and surgery.
Published at: July 31, 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.