MedPath

Irinotecan Advanced Drug Monograph

Published:Jul 16, 2025

Generic Name

Irinotecan

Brand Names

Camptosar, Onivyde, Onivyde pegylated liposomal (previously known as Onivyde)

Drug Type

Small Molecule

Chemical Formula

C33H38N4O6

CAS Number

97682-44-5

Associated Conditions

Esophageal Cancer, Ewing's Sarcoma, Gastric Cancer, High Grade Glioma: Glioblastoma (GBM), Metastatic Cervical Cancer, Non-Small Cell Lung Cancer (NSCLC), Ovarian Cancer, Pancreatic Cancer, Rhabdomyosarcomas, Small Cell Lung Cancer (SCLC), Stage IV Colorectal Cancer, Recurrent, metastatic Colorectal carcinoma, Refractory, metastatic Pancreatic adenocarcinoma

A Comprehensive Monograph on the Topoisomerase I Inhibitor Irinotecan

I. Executive Summary and Drug Profile

1.1. Overview of Irinotecan: A Semisynthetic Camptothecin Analog

[Irinotecan is a cornerstone chemotherapeutic agent in modern oncology, particularly in the management of gastrointestinal malignancies. It is classified pharmacologically as a topoisomerase I inhibitor, belonging to the camptothecin class of antineoplastic drugs.][1] Structurally, irinotecan is a semisynthetic, water-soluble derivative of camptothecin, a cytotoxic, quinoline-based alkaloid originally extracted from the Chinese ornamental tree,

Camptotheca acuminata[.][1][ This chemical modification to create a water-soluble analog was a critical step in overcoming the poor solubility and formulation challenges of the parent compound, thereby enabling its clinical development and intravenous administration.][3]

[A fundamental characteristic of irinotecan is its nature as a prodrug. It is pharmacologically inert in its administered form and requires in vivo metabolic activation to exert its cytotoxic effects.][1][ This conversion process, which occurs primarily in the liver and plasma, is central to its entire pharmacokinetic and pharmacodynamic profile. The reliance on metabolic activation introduces significant layers of complexity, including high interindividual variability in drug exposure, a distinct toxicity profile, and a susceptibility to numerous drug-drug interactions and pharmacogenomic influences, all of which are critical considerations in its clinical application.][7]

1.2. Chemical and Physical Properties

[Irinotecan is rigorously defined by its chemical and physical identifiers, which are essential for its synthesis, formulation, and regulatory classification. The unique Chemical Abstracts Service (CAS) Registry Number for the free base form is 97682-44-5, and its DrugBank accession number is DB00762.][1][ It is also identified by other related CAS numbers for its salt and impurity forms, such as 100286-90-6 for the hydrochloride (HCl) salt and 136572-09-3 for the HCl trihydrate, which is often the form used in clinical preparations.][5]

[The molecular formula of irinotecan free base is C33​H38​N4​O6​, corresponding to a molecular weight of approximately 586.68 g/mol.][9][ Its systematic IUPAC name is (S)-4,11-diethyl-4-hydroxy-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3',4':6,7]indolizino[1,2-b]quinolin-9-yl [1,4'-bipiperidine]-1'-carboxylate, which describes its complex pentacyclic ring structure derived from camptothecin with a bis-piperidine side chain responsible for its water solubility.][5][ Common synonyms used in research and clinical literature include CPT-11 and Camptothecin-11.][4]

[Physically, irinotecan is a pale yellow solid with a melting point of 222-223°C.][5][ It is a hydrophilic compound that is soluble in dimethyl sulfoxide (DMSO) and slightly soluble in other organic solvents such as acetonitrile and methanol when heated.][1] These properties dictate the requirements for its formulation as an injectable solution for intravenous administration.

1.3. Formulations and Brand Names

[Irinotecan is available globally in two principal formulations, which possess distinct pharmacokinetic properties and clinical applications. The first is the conventional formulation of irinotecan hydrochloride, most widely known by the brand name ]Camptosar[.][1] The second is a novel formulation,

Onivyde[, which consists of irinotecan hydrochloride trihydrate encapsulated within pegylated liposomal nanoparticles.][13][ Other international brand names for conventional irinotecan include Campto and Irinomedac.][4]

[The development and existence of these two formulations represent a deliberate therapeutic strategy to enhance the clinical utility of the drug. Conventional irinotecan (Camptosar) is associated with a challenging pharmacokinetic profile, characterized by rapid systemic clearance and significant, often dose-limiting, toxicities.][1][ The liposomal formulation (Onivyde) was engineered as a direct application of nanomedicine to address these limitations.][16][ The liposomal vesicle serves as a protective carrier for the irinotecan payload. This encapsulation shields the drug from premature metabolic conversion in the plasma, significantly prolongs its circulation half-life, and is designed to leverage the Enhanced Permeability and Retention (EPR) effect for preferential accumulation within tumor tissues.][17][ This fundamental difference in drug delivery technology results in a markedly different pharmacokinetic profile for Onivyde compared to Camptosar, which in turn alters its efficacy and safety profile. This distinction has led to separate regulatory approvals and a distinct clinical positioning for Onivyde, particularly in the treatment of pancreatic cancer, where it has established a new standard of care.][13]

[Major pharmaceutical companies involved in the manufacturing and distribution of irinotecan include Pfizer, which markets Camptosar, and Ipsen Biopharmaceuticals, Inc., which markets Onivyde.][20][ With the expiration of initial patents, generic versions of conventional irinotecan hydrochloride injection are also available from manufacturers such as Teva Pharmaceuticals.][22]

II. Molecular Pharmacology and Mechanism of Action

2.1. Prodrug Activation: The Critical Conversion of Irinotecan to SN-38

[The cytotoxic activity of irinotecan is entirely dependent on its biotransformation from an inactive prodrug to a highly potent active metabolite. Following intravenous administration, irinotecan undergoes extensive metabolic conversion, primarily through hydrolysis of the carbamate ester linkage at the C-10 position.][1][ This reaction is catalyzed by carboxylesterase (CES) enzymes, with CES2, predominantly found in the liver and intestines, exhibiting a significantly higher affinity and catalytic efficiency for irinotecan than the CES1 isoform.][1][ In addition to tissue-based conversion, hydrolysis also occurs in the plasma, mediated by butyrylcholinesterase (hBChE).][1]

[This metabolic activation yields the compound 7-ethyl-10-hydroxycamptothecin, universally known as SN-38.][1][ The pharmacological significance of this conversion cannot be overstated. In vitro cytotoxicity assays have consistently demonstrated that SN-38 is 100 to 1,000 times more potent as an inhibitor of topoisomerase I than its parent compound, irinotecan.][5][ Consequently, the formation of SN-38 is the rate-limiting step for irinotecan's therapeutic effect, and the systemic and intratumoral concentrations of SN-38 are the most relevant pharmacodynamic markers for both antitumor activity and toxicity.][1] The efficiency of this conversion, which displays high interindividual variability, is a primary determinant of patient outcomes.

2.2. The Ternary Complex: Inhibition of Topoisomerase I and Induction of DNA Lesions

[The molecular target of irinotecan's active metabolite, SN-38, is the nuclear enzyme DNA topoisomerase I (Topo I).][1][ Topo I plays an essential role in normal cell physiology by managing DNA topology. During critical cellular processes like DNA replication and transcription, the enzyme relieves the torsional strain that builds up in the DNA double helix by inducing transient, reversible single-strand breaks.][3] Once the strain is relieved, the enzyme re-ligates the broken DNA strand, allowing the process to continue.

[SN-38 exerts its cytotoxic effect by interrupting this catalytic cycle. It does not bind to the free enzyme or to DNA alone; instead, it specifically intercalates into the enzyme-DNA covalent complex at the site of the single-strand break.][1][ This binding stabilizes what is known as the "cleavable complex," creating a stable ternary structure of Topo I-DNA-SN-38 and physically preventing the enzyme from completing its function of re-ligating the DNA strand.][3] This action effectively "poisons" the enzyme, trapping it on the DNA.

[The formation of these stabilized ternary complexes is the initial DNA lesion. However, the ultimate lethal event occurs during the S-phase of the cell cycle, when the advancing DNA replication fork collides with these complexes.][3][ This collision converts the transient single-strand break into a permanent, irreversible, and highly cytotoxic double-strand DNA break.][3][ This mechanism explains why irinotecan is a cell cycle-specific agent, exerting its maximal effect on rapidly proliferating cells that are actively synthesizing DNA.][3]

2.3. Induction of Apoptosis and Cell Cycle Arrest

[The accumulation of extensive and irreparable double-strand DNA breaks serves as a powerful damage signal within the cancer cell. This signal activates complex intracellular damage response pathways, leading to two primary outcomes: cell cycle arrest and apoptosis.][29][ The cell cycle is halted, typically in the S and G2 phases, to prevent the propagation of damaged genetic material.][29][ If the DNA damage is too severe to be repaired, the cell is directed to undergo programmed cell death, or apoptosis.][11][ While the tumor suppressor protein p53 is a key mediator in this apoptotic cascade, evidence also suggests the existence of p53-independent pathways through which irinotecan can induce cell death, which is relevant for treating tumors with mutated or non-functional p53.][33] The induction of apoptosis is the final step in irinotecan's mechanism of action and the ultimate basis of its antitumor activity.

[A distinct pharmacological action of irinotecan, unrelated to its primary cytotoxic mechanism, is responsible for the acute cholinergic syndrome observed in some patients. This syndrome, which includes symptoms like early-onset diarrhea, rhinitis, diaphoresis (sweating), and miosis (pupil constriction), is a direct consequence of the parent compound, irinotecan, inhibiting the enzyme acetylcholinesterase.][3][ This inhibition leads to an excess of acetylcholine at nerve synapses, causing a transient cholinergic crisis. This effect occurs during or shortly after the drug infusion and is mechanistically separate from the delayed-onset diarrhea caused by the SN-38 metabolite's toxicity to the gastrointestinal mucosa. The distinct etiology of the early cholinergic symptoms explains why they are effectively prevented or treated with a competitive antagonist of acetylcholine, such as atropine.][26] This differentiation is crucial for appropriate clinical management of the drug's side effects.

III. Clinical Pharmacokinetics and Metabolism

The clinical pharmacokinetics of irinotecan are exceptionally complex, characterized by a multi-compartmental distribution, extensive metabolic conversion involving a delicate balance of activation and detoxification, a pH-dependent equilibrium, and significant interindividual variability. Understanding these processes is paramount for optimizing therapy and managing toxicity.

3.1. Administration, Distribution, and Protein Binding

[Irinotecan is administered exclusively by intravenous (IV) infusion. Conventional irinotecan (Camptosar) is typically infused over 30 to 90 minutes, while the liposomal formulation (Onivyde) is administered as a 90-minute IV infusion.][1] Following administration, irinotecan, a hydrophilic compound, exhibits a large volume of distribution at steady state (

[Vss​), with reported values ranging from 110 to 445 L/m2, indicating extensive distribution into peripheral tissues.][1][ The drug has been detected in various bodily fluids, including pleural fluid, sweat, and saliva, underscoring its wide dissemination.][3]

[Plasma protein binding differs substantially between the parent drug and its active metabolite. Irinotecan itself is moderately bound to plasma proteins, with bound fractions ranging from 30% to 68%.][3][ In stark contrast, the active metabolite SN-38 is highly protein-bound, with approximately 95% associated with plasma proteins, primarily albumin.][1][ This high degree of binding for SN-38 effectively creates a circulating reservoir of the active drug, influencing its disposition, prolonging its effective half-life, and stabilizing its active lactone configuration.][1]

3.2. The Lactone-Carboxylate Equilibrium: A pH-Dependent Dynamic

[A critical and dynamic aspect of irinotecan's pharmacology is the reversible, pH-dependent equilibrium between two distinct chemical forms. Both irinotecan and SN-38 possess an α-hydroxy-δ-lactone ring (the E-ring of the camptothecin structure), which is essential for binding to the Topo I-DNA complex and exerting cytotoxic activity.][8][ In an acidic environment (low pH), this closed-ring lactone form is favored. However, at neutral or physiological pH (e.g., plasma pH of ~7.4), the lactone ring undergoes spontaneous and reversible hydrolysis to open into an inactive, water-soluble carboxylate form.][1]

[The interconversion for irinotecan is rapid, with an equilibration half-life of approximately 14 minutes, whereas the process is slower for SN-38.][27][ In plasma, the equilibrium favors the inactive carboxylate form for the parent drug, but the active lactone form for the metabolite SN-38, partly due to the latter's preferential binding to albumin, which stabilizes the lactone ring.][1] This dynamic equilibrium has profound clinical implications. Any fluctuation in systemic or local pH can alter the concentration of the active drug form. While the physiological pH of blood favors the inactive form, the often-acidic microenvironment of solid tumors, a result of anaerobic glycolysis (the Warburg effect), could theoretically shift the equilibrium back toward the active lactone form upon drug accumulation in the tumor. This phenomenon might contribute to a degree of tumor-selective activation, although this has not been definitively proven as a major mechanism of action in humans.

3.3. The Metabolic Cascade: A Balance of Activation and Detoxification

[The net therapeutic and toxic effects of irinotecan are determined by a complex metabolic network that balances bioactivation with detoxification.][7] This network is the primary source of the drug's high interpatient variability and its susceptibility to pharmacogenomic and drug-drug interactions.

  • Activation Pathway:[ As previously described, the key activation step is the conversion of the irinotecan prodrug to the active metabolite SN-38. This is mediated primarily by the carboxylesterase CES2 in the liver and intestinal tissues.][1]
  • Detoxification Pathways: There are two main routes of detoxification:
  1. Glucuronidation of SN-38:[ The highly potent SN-38 is detoxified via conjugation with glucuronic acid to form the inactive and water-soluble metabolite, SN-38 glucuronide (SN-38G). This reaction is catalyzed almost exclusively by the enzyme UDP-glucuronosyltransferase 1A1 (UGT1A1) in the liver.][1] The activity of UGT1A1 is a critical determinant of SN-38 clearance and is the basis for the most significant pharmacogenomic interactions.
  2. Oxidation of Irinotecan:[ The parent drug, irinotecan, can also be metabolized to inactive compounds via oxidation. This pathway is mediated by the cytochrome P450 enzyme CYP3A4, which forms two primary metabolites: 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino]-carbonyloxycamptothecin (APC) and 7-ethyl-10-[4-amino-1-piperidino]-carbonyloxycamptothecin (NPC).][6] These metabolites have negligible topoisomerase I inhibitory activity.

The clinical outcome for a given patient depends on the delicate balance between the rate of CES-mediated activation and the rates of UGT1A1- and CYP3A4-mediated detoxification.

3.4. Excretion and Enterohepatic Recirculation

[The elimination of irinotecan and its metabolites from the body occurs through both renal and biliary routes.][3][ Urinary excretion accounts for approximately 11-20% of the parent drug, but less than 1% of SN-38 and 3% of SN-38G.][3][ The primary route of elimination is biliary excretion into the gastrointestinal tract.][3]

[A crucial event occurs once the inactive SN-38G metabolite is secreted into the gut lumen via the bile. Here, it encounters β-glucuronidase enzymes, which are produced in abundance by the commensal intestinal microflora.][8][ These bacterial enzymes cleave the glucuronide moiety from SN-38G, regenerating the highly active and toxic SN-38 directly within the intestinal tract. This locally regenerated SN-38 has two fates: it can be reabsorbed back into the systemic circulation, a process known as enterohepatic recirculation, or it can exert a direct toxic effect on the intestinal mucosa.][1]

[This enterohepatic recirculation is responsible for creating a characteristic "second peak" or rebound in the plasma concentration-time curve of SN-38, which occurs hours after the initial infusion peak.][1] More importantly, the direct exposure of the gut epithelium to high concentrations of regenerated SN-38 is the primary mechanism underlying the severe, dose-limiting, and potentially life-threatening late-onset diarrhea that is the hallmark toxicity of irinotecan therapy. This highlights the gut microbiome as a critical, non-host factor that profoundly influences the drug's safety profile.

Table 1: Summary of Key Pharmacokinetic Parameters of Irinotecan and SN-38

ParameterIrinotecanSN-38 (Active Metabolite)Source(s)
AdministrationIntravenous infusion (30-90 min)Formed in vivo1
Time to Peak Plasma Conc. (Tmax​)End of infusion~1-2 hours post-infusion3
Plasma Protein Binding30-68% (moderate)~95% (high), mainly to albumin3
Volume of Distribution (Vdss​)Large (e.g., 110-445 L/m2)Extensive tissue distribution3
Terminal Half-life (t1/2​)~6-14 hours~10-21 hours (longer than parent)3
Clearance (Total Body)~13-21 L/h/m2Lower than parent drug3
Metabolism- Activation by CES1/2, hBChE to SN-38 - Inactivation by CYP3A4 to APC/NPC- Inactivation by UGT1A1 to SN-38G1
ExcretionBiliary and RenalPrimarily biliary (as SN-38G), then fecal3
Key Pharmacokinetic FeatureHigh interpatient variability- High interpatient variability - Enterohepatic recirculation3

IV. Pharmacogenomics: UGT1A1-Guided Therapy

The substantial interindividual variability in irinotecan's toxicity profile is, to a significant extent, explained by pharmacogenomics. Genetic polymorphisms in the gene encoding the primary SN-38 detoxifying enzyme, UGT1A1, are the most well-characterized and clinically relevant predictors of severe adverse reactions to irinotecan.

4.1. UGT1A1 Polymorphisms (*28, *6) and Reduced Enzyme Activity

[The ]UGT1A1[ gene is highly polymorphic. Certain common genetic variants, known as star (*) alleles, result in reduced expression or functional activity of the UGT1A1 enzyme.][28] The most studied and clinically important of these is the

UGT1A1*28[ allele (also known as UGT1A1(TA)7), which is characterized by an extra 'TA' dinucleotide repeat in the TATA box of the gene's promoter region. This leads to reduced gene transcription and, consequently, lower enzyme levels.][37] Another important variant, particularly prevalent in Asian populations, is the

UGT1A1*6[ allele, a missense mutation in exon 1 that results in a functionally deficient enzyme.][28]

[Based on their ]UGT1A1 genotype, individuals can be classified into different metabolizer phenotypes:

  • Normal Metabolizers (NMs): Individuals with two normal function alleles (e.g., *1/*1).
  • Intermediate Metabolizers (IMs): Individuals heterozygous for a decreased function allele (e.g., *1/*28 or *1/*6).
  • Poor Metabolizers (PMs):[ Individuals homozygous for a decreased function allele (e.g., *28/*28 or *6/*6) or compound heterozygous for two different decreased function alleles (e.g., *6/*28).][28]

4.2. Clinical Impact: Increased Risk of Severe Neutropenia and Diarrhea

[The clinical consequence of reduced UGT1A1 activity is a decreased ability to metabolize and detoxify the active metabolite, SN-38. This leads to higher and more prolonged systemic exposure to SN-38 following irinotecan administration.][37][ As a direct result, patients who are UGT1A1 poor metabolizers (PMs) are at a significantly increased risk of developing severe (Grade 3-4) and potentially life-threatening toxicities, most notably neutropenia and diarrhea.][1][ Intermediate metabolizers (IMs) may also face an elevated risk compared to normal metabolizers, although generally to a lesser extent than PMs.][37] This strong, mechanistically plausible gene-drug interaction provides a compelling rationale for pre-treatment genotyping to identify at-risk patients and implement risk mitigation strategies, such as prophylactic dose reduction.

4.3. Genotype-Based Dosing Guidelines: A Comparative Analysis

[The robust evidence linking ]UGT1A1 genotype to irinotecan toxicity has led major regulatory agencies and pharmacogenetics consortia to issue specific dosing recommendations. While there is a broad international consensus on the principle of dose reduction for poor metabolizers, the specific recommendations and their strength vary.

  • U.S. Food and Drug Administration (FDA):[ The FDA-approved prescribing information for both conventional irinotecan (Camptosar) and liposomal irinotecan (Onivyde) includes recommendations for patients with reduced UGT1A1 activity. The labels state that a reduction in the starting dose should be ]considered[ for patients known to be homozygous for the ]UGT1A1*28[ allele (the Camptosar label also includes the ]UGT1A1*6[ allele).][37][ For Onivyde, the label provides a specific recommendation: the starting dose in homozygous] UGT1A1*28[ patients should be 50 mg/m², which can be escalated to the standard 70 mg/m² in subsequent cycles if the initial dose is well tolerated.][37] The FDA's language ("consider") provides guidance but does not mandate pre-treatment testing.
  • European Medicines Agency (EMA):[ The EMA's guidance for Onivyde aligns closely with the FDA's. The Summary of Product Characteristics recommends a reduced starting dose of 50 mg/m² for patients known to be homozygous for the ]UGT1A1*28[ allele, with the option to increase the dose to 70 mg/m² in subsequent cycles based on individual tolerance.][44]
  • Dutch Pharmacogenetics Working Group (DPWG):[ The DPWG has issued one of the strongest recommendations globally. They classify pre-treatment ]UGT1A1[ genotyping as "]essential[" for patient safety prior to initiating irinotecan.][28][ For patients identified as UGT1A1 poor metabolizers (PMs), the DPWG recommends a specific dose reduction:] start with 70% of the normal dose[. The dose can then be titrated upwards in subsequent cycles, guided by the patient's neutrophil count and overall tolerance.][28][ For intermediate metabolizers (IMs), the DPWG recommends no initial dose adjustment, reasoning that standard irinotecan dosing has largely been established in Western populations where the IM phenotype is common.][40]
  • Clinical Pharmacogenetics Implementation Consortium (CPIC):[ While a dedicated CPIC guideline for irinotecan and ]UGT1A1[ is not yet available, CPIC has formally defined the functionality of key ]UGT1A1[ alleles (*1 as normal function, *28 as decreased function).][28][ The principles outlined by CPIC align with other international guidelines, supporting the need for dose reduction in patients with two decreased-function alleles (i.e., poor metabolizers).][43][ The existing CPIC guideline for atazanavir and] UGT1A1[ provides the methodological framework and evidence grading system that would be applied to irinotecan.][47]

Table 2: Summary of UGT1A1 Genotype-Guided Dosing Recommendations

UGT1A1 Phenotype (Genotype Example)FDA (Camptosar) 37FDA (Onivyde) 37EMA (Onivyde) 44DPWG 28
Normal Metabolizer (NM) (e.g., *1/*1)Standard doseStandard dose (70 mg/m²)Standard dose (70 mg/m²)Standard dose
Intermediate Metabolizer (IM) (e.g., *1/*28, *1/*6)Increased risk of neutropenia noted. Closely monitor.Increased risk noted. Closely monitor.Increased risk noted. Closely monitor.No initial dose adjustment recommended.
Poor Metabolizer (PM) (e.g., *28/*28, *6/*6, *6/*28)Consider a reduction in the starting dose by at least one level.Recommended starting dose of 50 mg/m². Increase to 70 mg/m² as tolerated.Consider a reduced starting dose of 50 mg/m². Increase to 70 mg/m² as tolerated.Essential to reduce starting dose to 70% of standard. Increase dose as tolerated, guided by neutrophil count.

V. Clinical Efficacy in Approved Indications

Irinotecan has secured a pivotal role in the treatment of several solid tumors, with its most prominent approvals in metastatic colorectal and pancreatic cancers. Its efficacy is primarily demonstrated within combination chemotherapy regimens.

5.1. Metastatic Colorectal Cancer (mCRC)

[Metastatic colorectal cancer represents the primary and longest-standing indication for irinotecan. It is approved by regulatory bodies worldwide for use in both the first-line and second-line settings for patients with advanced disease.][1]

  • First-Line Therapy:[ Irinotecan is a component of standard first-line therapy, typically in combination with 5-fluorouracil (5-FU) and leucovorin (LV).][51][ The most widely used regimen is] FOLFIRI[, which consists of infusional 5-FU, LV, and irinotecan.][1][ Another established combination is] XELIRI[, which substitutes the oral fluoropyrimidine capecitabine for 5-FU/LV.][1]
  • Second-Line Therapy:[ Irinotecan is also indicated for patients whose disease has recurred or progressed following an initial fluorouracil-based therapy.][12] In this setting, it can be used as a single agent or as part of a combination regimen.

[The clinical development of irinotecan in mCRC has been characterized by numerous trials exploring its use in combination with targeted therapies to enhance efficacy. These include combinations with anti-VEGF agents like bevacizumab (e.g., FOLFIRI-bevacizumab) and anti-EGFR agents such as cetuximab and panitumumab for ]RAS[ wild-type tumors.][55] For example, the phase 3 IMPROVE trial investigated intermittent versus continuous FOLFIRI plus panitumumab in the first-line setting for

RAS/BRAF[ wild-type mCRC, demonstrating that an intermittent strategy was feasible and associated with reduced toxicity.][58] These combination strategies underscore that irinotecan's maximum benefit in mCRC is realized as part of a multi-drug approach.

5.2. Metastatic Pancreatic Adenocarcinoma (mPDAC)

The approval of irinotecan, specifically its liposomal formulation (Onivyde), has marked a significant advancement in the treatment paradigm for metastatic pancreatic adenocarcinoma, a disease with a historically poor prognosis.

  • Second-Line Therapy (Post-Gemcitabine):[ The initial approval for liposomal irinotecan was for the treatment of patients with mPDAC whose disease had progressed following gemcitabine-based chemotherapy.][17][ This approval was based on the pivotal, international, randomized phase 3] NAPOLI-1[ trial (NCT01494506).][60][ In this study, the combination of liposomal irinotecan plus 5-FU/LV demonstrated a statistically significant and clinically meaningful improvement in median overall survival (OS) compared to 5-FU/LV alone (6.1 months vs. 4.2 months, respectively; hazard ratio 0.67).][15][ Final analysis with extended follow-up confirmed this survival benefit (median OS 6.2 months vs. 4.2 months).][62]
  • First-Line Therapy (NALIRIFOX):[ In a landmark development in 2024, both the FDA and EMA approved liposomal irinotecan for the first-line treatment of mPDAC.][13][ This approval was for the] NALIRIFOX[ regimen, which combines liposomal irinotecan, oxaliplatin, 5-FU, and leucovorin.][13][ The approval was based on the results of the randomized, active-controlled, phase 3] NAPOLI-3[ trial (NCT04083235).][13][ This trial compared NALIRIFOX to the established standard-of-care regimen of gemcitabine plus nab-paclitaxel in 770 treatment-naïve patients with mPDAC.][13] The trial met its primary and key secondary endpoints, demonstrating a statistically significant superiority for NALIRIFOX:
  • Overall Survival (OS):[ Median OS was 11.1 months for the NALIRIFOX arm versus 9.2 months for the gemcitabine + nab-paclitaxel arm (HR 0.84; p=0.0403).][13]
  • Progression-Free Survival (PFS):[ Median PFS was 7.4 months for the NALIRIFOX arm versus 5.6 months for the control arm (HR 0.70; p=0.0001).][13]

[The NAPOLI-3 results established NALIRIFOX as a new standard-of-care first-line treatment option for mPDAC, representing the first phase 3 trial to demonstrate a survival benefit over the gemcitabine/nab-paclitaxel combination in this setting.][14]

VI. Investigational Uses and the Clinical Trial Landscape

Beyond its established roles in colorectal and pancreatic cancer, irinotecan's broad-spectrum cytotoxic activity has prompted extensive investigation across a range of other malignancies. The clinical trial landscape is rich with studies exploring its efficacy as a monotherapy, in novel combinations, and in new formulations for difficult-to-treat cancers.

6.1. Small-Cell Lung Cancer (SCLC)

[Irinotecan, typically in combination with a platinum agent like cisplatin or carboplatin, has long been recognized as an active regimen in small-cell lung cancer.][1] However, its precise role in the modern era of chemoimmunotherapy is still being defined.

  • Conventional Irinotecan:[ Early phase 2 trials established the activity of irinotecan/platinum combinations in newly diagnosed extensive-stage SCLC.][67] The Japanese Clinical Oncology Group (JCOG) conducted a phase 3 trial (JCOG9511) which showed that irinotecan plus cisplatin improved survival over the standard etoposide plus cisplatin, leading to its establishment as a standard regimen in Japan.
  • Liposomal Irinotecan (Onivyde):[ More recently, the liposomal formulation has been investigated. The phase 3 ]RESILIENT[ trial (NCT03088813) compared second-line liposomal irinotecan monotherapy against topotecan, a standard second-line agent.][69][ The study did not meet its primary endpoint of improving overall survival. However, liposomal irinotecan demonstrated a doubling of the objective response rate (ORR) and a more favorable safety profile, with a reduced incidence of Grade ≥3 treatment-emergent adverse events compared to topotecan.][69] This suggests that while not superior in OS, liposomal irinotecan is an active and potentially better-tolerated option in this setting.
  • Novel Combinations:[ The most recent research, presented at major oncology conferences in 2024, focuses on combining irinotecan with novel agents. A phase 1b/2 trial (NCT02611024) evaluating the combination of ]lurbinectedin and irinotecan[ in patients with relapsed SCLC showed highly promising results. In patients with a chemotherapy-free interval >30 days, the combination yielded an ORR of 52.7%, a median OS of 12.7 months, and a manageable safety profile.][71][ These data support the ongoing phase 3 LAGOON trial (NCT05153239), which includes a lurbinectedin/irinotecan arm and may establish this combination as a new standard of care for relapsed SCLC.][71]

6.2. Breast Cancer

[Irinotecan's role in breast cancer is purely investigational, with activity demonstrated in heavily pre-treated patients with metastatic disease. Several phase 2 trials have explored its use, but it has not been incorporated into standard-of-care guidelines.][73]

  • Monotherapy:[ The CALGB 9844 trial (NCT00003351) was a randomized phase 2 study that compared two different schedules of single-agent irinotecan (weekly vs. every 3 weeks) in patients with refractory metastatic breast cancer, aiming to define its activity and toxicity profile in this population.][73]
  • Combination Therapy:[ Other phase 2 studies have evaluated irinotecan in combination with various agents, including standard cytotoxics like docetaxel and etoposide, as well as targeted therapies such as the EGFR inhibitor cetuximab (for metastatic triple-negative breast cancer) and the HER2-targeted antibody trastuzumab (for HER2-positive disease).][73][ A trial in China (NCT03562390) is currently evaluating irinotecan as a third-line or later therapy for patients who have progressed on anthracyclines and taxanes.][75] These studies suggest that irinotecan may have a niche role for patients who have exhausted standard options, but further research is needed to identify the optimal combination and patient population.

6.3. Glioblastoma (GBM) and Other CNS Malignancies

[Irinotecan has been of interest for treating high-grade gliomas like glioblastoma due to its ability to penetrate the blood-brain barrier.][76] Its use has been most prominent in the recurrent setting, almost always in combination with the anti-angiogenic agent bevacizumab.

  • Bevacizumab Combinations:[ A series of phase 2 trials in the late 2000s and early 2010s established the bevacizumab-irinotecan combination as an active regimen for recurrent grade III and IV gliomas. These studies reported encouraging radiographic response rates, ranging from 26% to as high as 63%, with acceptable toxicity.][76][ This led to the regimen being widely used, although its survival benefit over bevacizumab monotherapy was subsequently debated. A randomized phase 2 trial (NCT00433381) directly compared bevacizumab plus irinotecan to bevacizumab plus temozolomide in patients with recurrent GBM.][78]
  • More Recent Strategies:[ Current research is focused on intensifying treatment. A retrospective study explored a triple-drug regimen of temozolomide, bevacizumab, and irinotecan, which was found to be well-tolerated.][79][ A prospective phase 1 study (NCT05201326) is actively investigating the safety and efficacy of combining irinotecan and bevacizumab with re-irradiation for patients with recurrent GBM, aiming to provide a higher level of evidence for this multi-modal approach.][80]

6.4. Other Solid Tumors

[The broad activity of irinotecan has led to its investigation in a wide variety of other advanced solid tumors, typically in phase 1 or 2 studies to establish safety and preliminary efficacy signals for new combinations or formulations.][81]

  • Gastrointestinal Cancers:[ Beyond colorectal and pancreatic cancer, irinotecan has been studied in advanced gastric and gastroesophageal junction adenocarcinoma. The phase 3 ]RINDBeRG[ trial investigated the strategy of continuing the anti-angiogenic agent ramucirumab beyond progression by combining it with irinotecan. The trial found that while the combination improved PFS, it did not confer a significant OS benefit over irinotecan monotherapy.][82][ Other phase 2 studies have explored combinations like apatinib plus irinotecan in this setting.][85]
  • Rare Cancers:[ Irinotecan has been evaluated in less common malignancies, such as small intestine adenocarcinoma, in combination with oxaliplatin and capecitabine (NCT00433550).][86]
  • Pediatric Sarcomas:[ The combination of irinotecan and temozolomide is a standard regimen for relapsed Ewing sarcoma. The COG study AEWS1221 (NCT01864109) is investigating the addition of this combination to the standard first-line chemotherapy backbone for newly diagnosed patients, aiming to improve outcomes from the outset.][87]

Table 3: Summary of Efficacy from Pivotal and Key Investigational Clinical Trials

Cancer TypeTrial Name / IDPhaseLine of TherapyRegimen(s) StudiedKey Efficacy Endpoints & ResultsSource(s)
mPDACNAPOLI-3 (NCT04083235)IIIFirst-LineNALIRIFOX (nal-IRI + Oxaliplatin + 5-FU/LV) vs. Gemcitabine + nab-PaclitaxelmOS: 11.1 vs. 9.2 months (HR 0.84, p=0.0403) mPFS: 7.4 vs. 5.6 months (HR 0.70, p=0.0001)13
mPDACNAPOLI-1 (NCT01494506)IIISecond-Line (post-Gem)nal-IRI + 5-FU/LV vs. 5-FU/LV alonemOS: 6.1 vs. 4.2 months (HR 0.67, p=0.012) mPFS: 3.1 vs. 1.5 months (HR 0.56, p<0.001)59
SCLCRESILIENT (NCT03088813)IIISecond-LineLiposomal Irinotecan vs. TopotecanmOS: Not significantly different ORR: 44.0% vs. 21.6% (doubled response rate)69
SCLCNCT02611024Ib/IISecond-LineLurbinectedin + IrinotecanORR: 52.7% mOS: 12.7 months mPFS: 5.0 months71
mCRCIMPROVE (NCT03318122)IIFirst-Line (RAS/BRAF wt)Intermittent vs. Continuous FOLFIRI + Panitumumab12-mo PFS: 58.5% (Intermittent) vs. 45.7% (Continuous) mOS: 35.1 vs. 36.3 months (equivalent)58
Adv. Gastric CancerRINDBeRG (jRCTs051180187)IIIThird-Line (post-Ramucirumab)Ramucirumab + Irinotecan vs. Irinotecan alonemOS: 9.4 vs. 8.5 months (HR 0.91, p=0.49) mPFS: 3.8 vs. 2.8 months (HR 0.72, p=0.002)82
Recurrent GBMVredenburgh et al. (NCT00317341)IIRecurrentBevacizumab + Irinotecan6-month PFS: 38% Radiographic Response Rate: 63%76

VII. Liposomal Irinotecan (Onivyde): A Comparative Analysis

The development of liposomal irinotecan (nal-IRI, Onivyde) represents a significant milestone in the application of nanomedicine to enhance the therapeutic index of a proven cytotoxic agent. Its properties, clinical data, and cost must be analyzed in comparison to conventional irinotecan.

7.1. Rationale and Pharmacological Advantages

[Liposomal irinotecan was engineered specifically to overcome the well-documented pharmacological limitations of the conventional, non-liposomal formulation.][17] The drug is encapsulated within a stable, long-circulating nanoliposomal particle, which confers several key advantages:

  • Prolonged Circulation:[ The pegylated liposome structure prevents rapid clearance by the reticuloendothelial system, significantly extending the systemic circulation time and plasma half-life of both irinotecan and its active metabolite, SN-38. Preclinical data suggest a 4-fold increase in half-life.][17]
  • Protection of Active Form:[ The liposome interior protects the irinotecan payload from premature hydrolysis to its inactive carboxylate form and from rapid conversion to SN-38 in the plasma. This ensures that a higher proportion of the drug remains in its active lactone configuration during circulation.][17]
  • Enhanced Tumor Delivery:[ The nanoparticle size allows nal-IRI to exploit the Enhanced Permeability and Retention (EPR) effect, leading to preferential accumulation in the leaky vasculature of tumor tissue. Macrophages within the tumor microenvironment are thought to play a role in taking up the liposomes and releasing the irinotecan payload locally, where it can be converted to SN-38.][16][ Preclinical models have shown up to a 50-fold higher drug exposure and a 5-fold higher SN-38 concentration in tumors with nal-IRI compared to conventional irinotecan.][18]

These pharmacological modifications are the mechanistic basis for nal-IRI's potential to improve efficacy and alter the toxicity profile compared to its conventional counterpart.

7.2. Comparative Efficacy and Safety

[While nal-IRI has a robust evidence base from the phase 3 NAPOLI trials in pancreatic cancer, its direct comparative efficacy against an active conventional irinotecan regimen (e.g., FOLFIRI) has not been established in a prospective, randomized setting.][89] The most informative data come from retrospective analyses.

[A key single-institution retrospective study compared nal-IRI/5FU with FOLFIRI in the second-line treatment of mPDAC.][89] Using inverse probability of treatment weighting to balance baseline characteristics, the study found:

  • Efficacy: There was no statistically significant difference in the primary outcome of progression-free survival (median PFS: 4.1 months for nal-IRI/5FU vs. 3.1 months for FOLFIRI) or in overall survival.
  • Safety and Tolerability:[ The frequency of adverse events was broadly similar. However, a notable difference was observed in the management of early-onset diarrhea. Patients receiving FOLFIRI required atropine for cholinergic symptoms significantly more often than those receiving nal-IRI/5FU (70% vs. 36%).][89] This finding supports the pharmacological rationale that the liposomal encapsulation shields the parent irinotecan drug, thereby reducing its direct anticholinesterase activity and mitigating the acute cholinergic syndrome.

7.3. The Cost-Effectiveness Dilemma

[The comparison between liposomal and conventional irinotecan introduces a significant health economics challenge. The same retrospective study that found similar efficacy also performed a cost analysis, concluding that the drug acquisition cost for a course of treatment with nal-IRI/5FU was nearly ]30 times more expensive[ than for FOLFIRI.][89]

This stark difference in cost, in the absence of prospectively proven superiority in efficacy, creates a complex decision-making landscape for clinicians, institutions, and payers. The choice between the two formulations may be influenced less by definitive clinical superiority and more by factors such as institutional budgets, insurance coverage, patient co-pays, and the perceived value of mitigating the risk of acute cholinergic symptoms. This scenario is a practical example of the broader debate in modern oncology regarding the value and cost-effectiveness of novel, high-cost formulations of existing drugs when they offer incremental or unproven benefits over older, less expensive standards.

7.4. Real-World Evidence (RWE)

As the use of Onivyde expands, real-world evidence is becoming increasingly important for validating the results of pivotal clinical trials in broader, more heterogeneous patient populations.

  • Efficacy in Clinical Practice:[ A real-world analysis presented in 2021 found that increased treatment duration with nal-IRI/5FU was associated with higher 1-year OS rates in patients with metastatic PDAC.][91][ A retrospective study from Belgium reported a median OS of 6.8 months and a median PFS of 3.1 months in a real-world cohort, figures that are consistent with the results of the NAPOLI-1 trial.][92]
  • Ongoing Research:[ Recognizing the need for region-specific data, a large real-world study (NCT07026123) is currently underway to evaluate the efficacy and safety of Onivyde-based regimens in Chinese patients with advanced pancreatic cancer.][93]

[This growing body of RWE is crucial for confirming the effectiveness and safety of liposomal irinotecan in routine clinical practice outside the controlled environment of a clinical trial and for understanding its long-term impact on patient outcomes.][66]

VIII. Safety Profile, Adverse Events, and Risk Management

Irinotecan is associated with a substantial and predictable toxicity profile, dominated by severe diarrhea and myelosuppression. Proactive monitoring and aggressive management of these adverse events are essential for patient safety and for maintaining the planned treatment schedule.

8.1. Comprehensive Adverse Event Profile

[The most common adverse reactions (reported in ≥30% of patients in clinical trials) across both monotherapy and combination regimens include a constellation of gastrointestinal and hematologic toxicities. These are nausea, vomiting, abdominal pain, diarrhea, constipation, anorexia, mucositis, neutropenia, leukopenia (including lymphocytopenia), anemia, and thrombocytopenia. Systemic symptoms such as asthenia (weakness), pain, fever, and alopecia (hair loss) are also very common.][1]

[Post-marketing pharmacovigilance, which captures adverse events in a much larger and more diverse patient population, has confirmed the expected toxicity profile but has also identified several rare or unexpected adverse drug event (ADE) signals. Analysis of the FDA Adverse Event Reporting System (FAERS) and the Japanese Adverse Drug Event Report (JADER) database has revealed significant associations between irinotecan and conditions such as second primary malignancies, hyperammonaemia, hiccups, significant skin toxicity, aphasia, and hepatic failure.][95][ The median time to onset for most ADEs is within the first month of treatment, emphasizing the need for close monitoring from the initiation of therapy.][95]

8.2. Boxed Warnings: A Detailed Analysis

[The U.S. Food and Drug Administration (FDA) has mandated a ]boxed warning[—its most serious type of warning—on the labels of both conventional (Camptosar) and liposomal (Onivyde) irinotecan. This warning highlights two potentially fatal toxicities: severe diarrhea and severe myelosuppression.][34]

  • Severe Diarrhea: The boxed warning explicitly distinguishes between two mechanistically different forms of diarrhea:
  • Early-Onset Diarrhea:[ Occurs during or shortly after the infusion (within 24 hours). It is part of a cholinergic syndrome and may be accompanied by symptoms such as rhinitis, increased salivation, and abdominal cramping. The warning notes that these symptoms can be ameliorated by the administration of atropine.][34]
  • Late-Onset Diarrhea:[ Occurs more than 24 hours after administration. This form can be prolonged, severe, and life-threatening, leading to dehydration, electrolyte imbalance, and sepsis. The warning mandates prompt and aggressive treatment with high-dose loperamide. It also stresses the need to monitor patients carefully, provide fluid and electrolyte replacement, and consider antibiotic support if ileus, fever, or severe neutropenia develops. If severe diarrhea occurs, irinotecan treatment must be interrupted and subsequent doses reduced.][34]
  • Severe Myelosuppression:[ The warning highlights that irinotecan can cause severe, life-threatening myelosuppression, primarily neutropenia. Deaths due to sepsis following severe neutropenia have been reported. This necessitates regular monitoring of blood counts and dose modification or interruption if severe neutropenia occurs.][34]

The presence of this boxed warning underscores that these are the two most critical, dose-limiting toxicities that every prescribing clinician must be prepared to anticipate and manage aggressively.

8.3. Contraindications, Warnings, and Precautions

Beyond the boxed warnings, the use of irinotecan is governed by several contraindications and precautions.

  • Contraindications:[ Irinotecan is absolutely contraindicated in patients with a history of a severe hypersensitivity reaction to irinotecan or any of its excipients.][42][ The label for Camptosar also lists chronic inflammatory bowel disease and/or bowel obstruction as contraindications.][96][ Due to the presence of irinotecan and its metabolites in breast milk, breastfeeding is contraindicated during treatment and for a period after the final dose.][2]
  • Warnings and Precautions:
  • Hypersensitivity:[ Severe anaphylactic or anaphylactoid reactions have been observed. The drug must be permanently discontinued if such a reaction occurs.][12]
  • Pulmonary Toxicity:[ Rare but potentially fatal cases of Interstitial Pulmonary Disease (IPD)-like events have been reported. Irinotecan should be interrupted in any patient who develops new or progressive dyspnea, cough, or fever, pending diagnostic evaluation. If IPD is confirmed, the drug should be permanently discontinued.][34]
  • Renal Impairment:[ Cases of renal impairment and acute renal failure have been identified, typically in patients who become severely volume-depleted from vomiting and/or diarrhea. It is not a direct nephrotoxin, but can cause renal failure secondary to its GI toxicity.][34]
  • Hepatic Impairment:[ Irinotecan clearance is diminished in patients with elevated bilirubin levels. Patients with total bilirubin between 1.0 and 2.0 mg/dL have a greater likelihood of developing Grade 3-4 neutropenia. The drug has not been formally studied in patients with bilirubin > 2.0 mg/dL, and its use in this population requires extreme caution.][34]
  • Embryo-Fetal Toxicity:[ Irinotecan is teratogenic and can cause fetal harm. Females of reproductive potential must be advised of the risk and use effective contraception during and for a significant period after treatment (e.g., 7 months for liposomal irinotecan). Male patients with female partners of reproductive potential must also use effective contraception (e.g., condoms) during and for months after treatment.][12]

8.4. Special Focus: Management of Irinotecan-Induced Diarrhea (IID)

[Given that diarrhea is the most characteristic and challenging toxicity of irinotecan, its management warrants a detailed focus. The dual mechanism—an acute cholinergic phase and a delayed, metabolite-driven secretory phase—requires a two-pronged management approach.][3]

  • Standard Management:
  • Early-Onset Diarrhea (<24 hours):[ This is managed with prophylactic or therapeutic administration of an anticholinergic agent, typically ]atropine[ at a dose of 0.25 mg to 1 mg administered intravenously or subcutaneously, unless clinically contraindicated.][26]
  • Late-Onset Diarrhea (>24 hours):[ This requires immediate and aggressive intervention. The standard of care is high-dose ]loperamide[. Patients should be instructed to take 4 mg (two capsules) at the first sign of a loose stool, followed by 2 mg (one capsule) every 2 hours until they have been diarrhea-free for at least 12 hours. During the night, the interval can be extended to 4 mg every 4 hours to reduce sleep disruption.][96][ It is critical that patients understand this regimen supersedes the standard over-the-counter dosing instructions for loperamide. For diarrhea that is refractory to high-dose loperamide, the somatostatin analog] octreotide[ is the recommended second-line agent.][102][ Throughout any episode of late-onset diarrhea, aggressive oral or intravenous fluid and electrolyte replacement is essential to prevent dehydration and its sequelae.][34]
  • Emerging and Investigational Strategies:[ The significant morbidity associated with IID has spurred research into alternative and prophylactic strategies, particularly for high-risk patients.][38] These emerging therapies target various points in the pathophysiological cascade:
  • Microbiome Modulation:[ Use of probiotics (e.g., ]Lactobacillus[ species) to alter the composition of the gut flora and reduce the activity of β-glucuronidase-producing bacteria.][105][ Prophylactic antibiotics, such as third-generation cephalosporins, have also been explored, particularly in pediatric protocols with protracted dosing schedules.][106]
  • Enzyme Inhibition:[ Development of specific inhibitors of bacterial β-glucuronidase to prevent the reactivation of SN-38 in the gut.][105]
  • Adsorbents:[ Use of oral carbonaceous adsorbents like activated charcoal (e.g., AST-120) to bind free SN-38 in the intestinal lumen and prevent mucosal damage.][106]
  • Traditional Chinese Medicine (TCM):[ Several herbal formulations, such as Huangqin Decoction and Hange-Shashin-To, have shown promise in both preclinical and clinical studies for their ability to reduce inflammation, protect the mucosal barrier, and alleviate IID.][102]

This shift towards prophylactic or pre-emptive management, especially in high-risk populations (e.g., UGT1A1 PMs) or with high-risk regimens, represents a key evolution in clinical practice, moving from reactive treatment to proactive risk mitigation.

Table 4: Clinical Management Algorithm for Irinotecan-Induced Diarrhea (IID)

Type of DiarrheaNCI CTCAE GradePatient Action & Clinical ManagementDose ModificationSource(s)
Early-Onset (During or <24h post-infusion)Any Grade- Inform clinical staff immediately. - Administer Atropine 0.25-1 mg IV/SC (unless contraindicated). - Monitor for cholinergic symptoms (cramping, sweating, rhinitis).Generally not required for early-onset diarrhea alone if it resolves quickly.26
Late-Onset (>24h post-infusion)Grade 1 (<4 stools/day over baseline)- Begin high-dose loperamide: 4 mg at first onset, then 2 mg q2h. - Maintain aggressive oral hydration. - Inform physician.Maintain dose level for next cycle.36
Grade 2 (4-6 stools/day over baseline)- Continue high-dose loperamide (2 mg q2h). - Continue aggressive oral hydration. - Contact physician for assessment.Maintain dose level for next cycle.36
Grade 3 (≥7 stools/day over baseline; incontinence; hospitalization indicated)- Medical emergency. - Hospitalize for IV fluids and electrolyte replacement. - Continue high-dose loperamide. - Consider adding octreotide if no improvement in 24-48h. - Obtain stool cultures; consider antibiotics if febrile or neutropenic.Interrupt therapy. Upon recovery, decrease irinotecan dose by one level (e.g., 25-50 mg/m²) for the next cycle.34
Grade 4 (Life-threatening consequences; urgent intervention indicated)- Medical emergency. - Immediate hospitalization and intensive supportive care (IV fluids, electrolytes, antibiotics). - Continue high-dose loperamide and/or octreotide.Interrupt therapy. Upon recovery, decrease irinotecan dose by at least one to two levels (e.g., 50 mg/m²) for the next cycle.34

IX. Drug-Drug and Drug-Disease Interactions

The complex metabolism and significant toxicity profile of irinotecan make it susceptible to a wide range of clinically important interactions with other drugs and pre-existing patient conditions. Vigilant medication reconciliation and patient assessment are mandatory to ensure safety.

9.1. Interactions Affecting Metabolism (CYP3A4 and UGT1A1 Modulators)

[The most significant drug-drug interactions are pharmacokinetic in nature, involving drugs that modulate the activity of the key metabolizing enzymes, CYP3A4 and UGT1A1.][7]

  • Strong CYP3A4 and/or UGT1A1 Inhibitors:[ Co-administration of potent inhibitors of these enzymes can significantly decrease the clearance of irinotecan and its active metabolite SN-38, leading to increased systemic exposure and a heightened risk of severe toxicity. It is strongly recommended to ]avoid the concurrent use of irinotecan with these agents. Examples include:
  • Azole antifungals:[ Ketoconazole, itraconazole.][12]
  • HIV Protease Inhibitors:[ Atazanavir, ritonavir, indinavir, lopinavir.][12]
  • Macrolide Antibiotics:[ Clarithromycin, erythromycin.][107]
  • Other Inhibitors:[ Gemfibrozil (a UGT1A1 inhibitor), nefazodone, cobicistat.][12]
  • [If use is unavoidable, strong CYP3A4 inhibitors should be discontinued at least one week prior to starting irinotecan therapy.][12]
  • Strong CYP3A4 Inducers:[ Conversely, co-administration with potent inducers of CYP3A4 can accelerate the metabolism of irinotecan, leading to substantially lower plasma concentrations of both the parent drug and SN-38. This carries the risk of reduced therapeutic efficacy. It is strongly recommended to ]avoid these combinations. If unavoidable, substitution with non-enzyme-inducing therapies should be made at least two weeks before initiating irinotecan. Examples include:
  • Anticonvulsants:[ Carbamazepine, phenytoin, phenobarbital.][12]
  • Rifamycins:[ Rifampin.][12]
  • Herbal Supplements:[ St. John's Wort.][12]
  • Dietary Interactions:[ Certain dietary components can also interact. Grapefruit juice is a known CYP3A4 inhibitor and should be avoided.][104][ Products containing turmeric (curcumin) may decrease irinotecan's effects and should also be avoided during treatment.][108]

9.2. Pharmacodynamic Interactions

Pharmacodynamic interactions occur when co-administered drugs have additive or synergistic effects on the body, independent of pharmacokinetic changes.

  • Myelosuppressive Agents: The risk of severe neutropenia and other cytopenias is increased when irinotecan is given with other drugs that suppress bone marrow function.
  • Laxatives:[ The concurrent use of laxatives should be avoided as it can exacerbate the risk and severity of irinotecan-induced diarrhea.][109]
  • Live Vaccines:[ Due to the immunosuppressive effects of irinotecan, administration of live or live-attenuated vaccines is contraindicated. The patient's immune response to the vaccine may be diminished, and there is a risk of developing a disseminated infection from the vaccine strain.][104]

9.3. Disease-State Interactions

[The use of irinotecan must be carefully considered in patients with certain pre-existing medical conditions, as these can significantly increase the risk of severe toxicity.][110]

  • Hepatic Dysfunction:[ As irinotecan is extensively metabolized in the liver, patients with hepatic impairment (particularly elevated bilirubin) have diminished drug clearance and increased exposure to SN-38, heightening the risk of severe hematologic toxicity. Caution is advised, and the drug has not been studied in patients with bilirubin > 2.0 mg/dL.][42][ Patients with Gilbert's syndrome, a condition of congenital UGT1A1 deficiency, are at a greater risk of myelosuppression.][104]
  • Renal Impairment:[ While the influence of renal impairment on irinotecan pharmacokinetics has not been fully evaluated, caution is recommended. Cases of acute renal failure have been reported, usually secondary to severe dehydration. The drug is not recommended for use in patients on dialysis.][42]
  • Myelosuppression:[ Patients with pre-existing bone marrow depression are at high risk for additive toxicity. Therapy should be administered cautiously, and close monitoring of blood counts is essential.][112]
  • Pulmonary Disease:[ Patients with pre-existing lung disease are considered a risk factor for developing the rare but fatal interstitial pulmonary disease (IPD) associated with irinotecan.][34]

Table 5: Clinically Significant Drug Interactions with Irinotecan

Interacting Agent / ClassMechanism of InteractionClinical ConsequenceManagement RecommendationSource(s)
Strong CYP3A4 Inducers (e.g., Carbamazepine, Phenytoin, Rifampin, St. John's Wort)Induction of CYP3A4 metabolismDecreased plasma concentrations of irinotecan and SN-38; potential for reduced efficacy.Avoid combination. Substitute with non-inducing therapy at least 2 weeks prior to starting irinotecan.12
Strong CYP3A4 Inhibitors (e.g., Ketoconazole, Clarithromycin, Ritonavir)Inhibition of CYP3A4 metabolismIncreased plasma concentrations of irinotecan and SN-38; increased risk of severe toxicity.Avoid combination. Discontinue inhibitor at least 1 week prior to starting irinotecan.12
Strong UGT1A1 Inhibitors (e.g., Atazanavir, Gemfibrozil)Inhibition of UGT1A1-mediated glucuronidation of SN-38Increased plasma concentrations of active SN-38; increased risk of severe toxicity (neutropenia, diarrhea).Avoid combination.12
Other Myelosuppressive AgentsAdditive pharmacodynamic effectIncreased severity and duration of neutropenia, thrombocytopenia, and anemia.Monitor blood counts closely. Consider dose reductions of one or both agents.112
LaxativesAdditive pharmacodynamic effectIncreased risk and severity of diarrhea.Avoid concurrent use.109
Live-Attenuated VaccinesPharmacodynamic antagonism (immunosuppression)Diminished vaccine efficacy and risk of disseminated infection from the vaccine virus.Contraindicated. Avoid for at least 3 months after cessation of therapy.107
Grapefruit Juice / TurmericCYP3A4 inhibition / Altered metabolismIncreased exposure (grapefruit) or decreased efficacy (turmeric).Avoid consumption during treatment.104

X. Mechanisms of Resistance and Strategies for Mitigation

Despite its efficacy, the development of therapeutic resistance is a major clinical challenge that limits the long-term benefit of irinotecan-based chemotherapy. Resistance is a multifactorial process involving alterations in drug transport, metabolism, target engagement, and the broader tumor ecosystem.

10.1. Upregulation of Drug Efflux Transporters

[A primary mechanism of acquired resistance to irinotecan is the overexpression of ATP-binding cassette (ABC) superfamily drug efflux transporters on the cancer cell membrane.][31] These transporters function as energy-dependent pumps that actively expel xenobiotics, including chemotherapeutic drugs, from the cell's interior. The most significant transporter implicated in irinotecan resistance is

ABCG2[, also known as Breast Cancer Resistance Protein (BCRP).][30][ Cancer cells that upregulate ABCG2 can efficiently pump out both irinotecan and its active metabolite SN-38, thereby lowering their intracellular concentrations below the threshold required for effective Topo I inhibition and cytotoxicity.][31] This is a classic mechanism of multidrug resistance and a significant barrier to successful treatment.

10.2. Alterations in Drug Metabolism and Target

Resistance can also develop through alterations at the level of intracellular drug metabolism or at the drug's molecular target, Topoisomerase I.

  • Metabolic Alterations:[ Cancer cells can adapt to increase the rate of SN-38 detoxification. This can occur through the induction of UGT1A1 expression within the tumor cell itself, leading to more rapid conversion of active SN-38 to inactive SN-38G.][31] Conversely, decreased expression or activity of the activating enzyme, CES2, could also lead to resistance by reducing the conversion of irinotecan to SN-38, although this is less commonly cited than efflux or detoxification mechanisms.
  • Target Alterations: Resistance can arise from changes to the Topoisomerase I enzyme itself. This can involve:
  • Quantitative changes:[ A decrease in the overall expression level of Topo I protein, resulting in fewer available targets for SN-38 to bind.][31]
  • Qualitative changes:[ The acquisition of missense mutations in the ]TOP1[ gene that alter the drug-binding site on the enzyme. Such mutations can reduce the binding affinity of SN-38, rendering the enzyme less susceptible to inhibition.][30]
  • [A recent study has also proposed a novel mechanism involving the gradual accumulation of mutations in non-coding DNA at Topo I cleavage sites, which reverts cleavage-sensitive sequences to cleavage-resistant ones, thereby reducing the chances of DNA break formation upon drug exposure.][114]

10.3. The Role of the Tumor Microenvironment and Cancer Stem Cells (CSCs)

Resistance to irinotecan is not solely a cell-autonomous phenomenon but is profoundly influenced by the complex tumor ecosystem.

  • Tumor Microenvironment (TME):[ The TME can promote resistance through various mechanisms, including the secretion of growth factors and cytokines that activate pro-survival signaling pathways in cancer cells. Furthermore, intercellular communication via extracellular vesicles (e.g., exosomes) can transfer resistance-conferring molecules, such as miRNAs or even the ABCG2 transporter protein itself, from resistant to sensitive cells.][31]
  • Cancer Stem Cells (CSCs):[ Tumors often contain a small, resilient subpopulation of cells known as cancer stem cells. These CSCs are characterized by their capacity for self-renewal and their intrinsic resistance to conventional chemotherapies, including irinotecan.][31][ CSCs often exhibit high levels of drug efflux pump expression and possess enhanced DNA damage repair capabilities. The survival of this CSC population following chemotherapy is believed to be a major driver of tumor relapse and metastasis.][31]

10.4. Clinical Strategies to Overcome Irinotecan Resistance

Overcoming irinotecan resistance is a key focus of clinical research. The primary strategy involves the use of rational combination therapies designed to target resistance mechanisms or complementary cellular pathways.

  • Targeting Parallel/Feedback Pathways:[ A common approach is to combine irinotecan with agents that block pro-survival signaling pathways that are activated as a resistance mechanism. For example, in ]RAS[ wild-type colorectal cancer, resistance to EGFR inhibitors can occur. Combining irinotecan with an EGFR inhibitor like cetuximab targets both Topo I and the EGFR pathway, a strategy proven to be effective after irinotecan failure.][31]
  • Investigational Approaches: Emerging strategies involve combining irinotecan with drugs that can re-sensitize resistant cells. These include:
  • Epigenetic Modifiers:[ Agents like DNA methyltransferase (DNMT) inhibitors are being explored to reverse epigenetic changes (e.g., demethylate the ]ABCG2[ promoter) that contribute to resistance.][31]
  • Signaling Pathway Inhibitors:[ Targeting pathways like NF-κB, which is implicated in pro-survival signaling and inflammation, has been shown in preclinical models to potentiate irinotecan's efficacy and may help overcome resistance.][33]
  • Novel Drug Combinations:[ As seen in SCLC, combining irinotecan with new classes of drugs, such as lurbinectedin, can produce synergistic effects and overcome prior resistance.][71]

The future of managing irinotecan resistance lies in a deeper understanding of the specific molecular drivers in individual patients, paving the way for personalized combination strategies.

XI. Future Perspectives and Novel Camptothecin Analogs

The clinical journey of irinotecan is far from over. Ongoing research is focused on both optimizing the use of the existing drug through smarter regimens and developing next-generation analogs with improved therapeutic properties.

11.1. Optimizing Irinotecan-Based Regimens

Current clinical research aims to enhance the therapeutic index of irinotecan by moving beyond simple dose escalation towards more sophisticated and sustainable treatment strategies.

  • Novel Combination Therapies:[ A major avenue of investigation is the combination of irinotecan with immunotherapy. The rationale is that the cytotoxic effect of irinotecan can induce immunogenic cell death, releasing tumor antigens and creating a more inflamed tumor microenvironment, which may synergize with the action of immune checkpoint inhibitors (e.g., anti-PD-1/PD-L1 antibodies). The IRICO study (jRCTs071210090) is a phase 2 trial prospectively evaluating irinotecan after progression on first-line chemo-immunotherapy in SCLC, which will provide crucial data on the efficacy of chemotherapy in the post-immunotherapy setting.][118][ Other studies are exploring combinations with novel agents like the alkylating agent lurbinectedin, which has shown remarkable synergy.][71]
  • Alternative Dosing Schedules:[ To mitigate toxicity and potentially delay the onset of resistance, alternative dosing schedules are being explored. The phase 2 IMPROVE trial in mCRC tested an intermittent FOLFIRI plus panitumumab schedule (8 cycles of induction followed by a treatment-free interval) against continuous treatment. The intermittent approach was found to be feasible, less toxic, and resulted in equivalent survival outcomes, suggesting that "drug holidays" may be a viable strategy to improve quality of life without compromising efficacy.][58]

11.2. The Next Generation: A Review of Novel Camptothecin Derivatives

[The success of irinotecan, coupled with its significant limitations, has fueled a decades-long search for superior camptothecin analogs. A wide array of novel derivatives are in various stages of preclinical and clinical development, each featuring chemical modifications designed to improve upon the parent compound's properties.][121] Key objectives of these modifications include enhancing water solubility, improving oral bioavailability, increasing the stability of the active lactone ring, boosting potency, and overcoming known resistance mechanisms like efflux pump overexpression.

[A review of some prominent analogs in development reveals the diversity of these approaches ][121]:

  • Rubitecan (9-nitrocamptothecin): An orally available analog that has shown activity in pancreatic cancer but was largely ineffective in other solid tumors.
  • Exatecan (DX-8951f): A potent, water-soluble, fully synthetic analog that does not require enzymatic activation. It showed moderate activity in breast and ovarian cancer but was not superior to gemcitabine in a phase 3 pancreatic cancer trial.
  • Gimatecan (ST1481): A highly lipophilic oral analog designed to increase cellular accumulation and overcome MDR-1/BCRP-mediated resistance. It has shown activity in early phase trials for glioma and colorectal cancer.
  • Belotecan (CKD-602): A water-soluble analog that has demonstrated promising response rates in phase 2 trials for ovarian cancer and SCLC, though with significant neutropenia.
  • Homocamptothecins (e.g., Diflomotecan): These analogs feature a more stable 7-membered lactone ring, which is less susceptible to pH-dependent hydrolysis, potentially reducing toxicity.
  • Polymer-Drug Conjugates (e.g., Prothecan, DE-310): These approaches link the camptothecin molecule to a polymer carrier (like PEG or dextran) to create a high-molecular-weight prodrug. This strategy aims to prolong circulation time and achieve selective tumor targeting via the EPR effect, similar to the principle behind liposomal irinotecan.

Despite the ingenuity of these chemical modifications, a persistent theme across the development of these analogs is the recurrence of myelosuppression and gastrointestinal toxicity as the primary dose-limiting toxicities. This pattern strongly suggests that these adverse effects are intrinsically linked to the on-target mechanism of topoisomerase I inhibition in rapidly dividing normal host tissues, such as bone marrow and the gastrointestinal epithelium. While molecular modifications can fine-tune potency and pharmacokinetics, they may be unable to fully uncouple the desired antitumor effect from the undesired toxicity in these tissues. This observation reinforces the importance of advanced drug delivery strategies, such as the liposomal encapsulation of Onivyde, which aim to physically sequester the drug away from sensitive normal tissues and preferentially deliver it to the tumor site. The future of this drug class may depend as much on innovations in delivery technology as on the discovery of new chemical entities.

XII. Conclusion and Recommendations for Clinical Practice

12.1. Synthesized View of Irinotecan's Role in Modern Oncology

Irinotecan has evolved from a promising but highly toxic plant-derived compound into an indispensable agent in the armamentarium against solid tumors, particularly those of the gastrointestinal tract. Its journey epitomizes several key themes in modern oncology: the power of chemical modification to create clinically viable drugs from natural products; the critical importance of understanding complex pharmacokinetics and metabolism; and the transformative potential of pharmacogenomics and nanomedicine to improve the therapeutic index of cytotoxic agents.

Initially hampered by a challenging toxicity profile, the clinical utility of irinotecan was unlocked through its integration into combination regimens like FOLFIRI, which remain a global standard of care for metastatic colorectal cancer. The subsequent elucidation of its metabolic pathway revealed the pivotal role of the UGT1A1 enzyme, leading to one of the most well-established examples of genotype-guided dosing in oncology. This allows for the prospective identification of patients at high risk for severe toxicity and the implementation of dose-reduction strategies to mitigate harm. Most recently, the development of liposomal irinotecan (Onivyde) has provided a powerful new formulation that leverages nanomedicine to optimize drug delivery, leading to a new first-line standard of care in metastatic pancreatic cancer—a landmark achievement in a notoriously difficult-to-treat disease. Irinotecan is no longer just a single drug, but a platform for therapeutic innovation, with ongoing research exploring novel combinations, schedules, and next-generation analogs.

12.2. Key Considerations for Clinicians: Dosing, Monitoring, and Toxicity Management

For the practicing oncologist, the safe and effective use of irinotecan requires a nuanced understanding of its properties and a commitment to proactive risk management. Based on the comprehensive evidence reviewed, the following actionable recommendations are critical for clinical practice:

  1. Embrace Pre-treatment UGT1A1 Genotyping: Given the strong evidence linking UGT1A1 poor metabolizer status to a significantly increased risk of severe, life-threatening neutropenia and diarrhea, pre-treatment genotyping should be considered standard practice. Adherence to genotype-guided dosing recommendations from regulatory bodies and pharmacogenetics consortia (e.g., FDA, EMA, DPWG) is essential for mitigating this predictable risk. For patients identified as poor metabolizers, initiating therapy at a reduced dose (e.g., a 30% reduction or as specified by the drug label) is a critical safety measure.
  2. Implement Aggressive, Protocol-Driven Management of Diarrhea: Irinotecan-induced diarrhea is a medical emergency that requires immediate and aggressive management. Clinical teams must have clear, established protocols for its treatment. This includes educating patients to distinguish between early- and late-onset diarrhea, ensuring they have access to high-dose loperamide, and providing explicit instructions for its use at the very first sign of a loose stool. Clinicians must have a low threshold for admitting patients with Grade 3/4 or refractory diarrhea for intravenous hydration, electrolyte management, and potential second-line therapy with octreotide.
  3. Conduct Vigilant Medication Reconciliation: The risk of severe toxicity is substantially increased by concurrent use of strong inhibitors of CYP3A4 or UGT1A1, while efficacy can be compromised by strong CYP3A4 inducers. A thorough review of all concomitant medications, including over-the-counter drugs (e.g., cimetidine) and herbal supplements (e.g., St. John's Wort, turmeric), is mandatory before every treatment cycle. High-risk combinations should be avoided, and alternative medications should be substituted whenever possible.
  4. Carefully Consider the Risk-Benefit-Cost Profile of Formulations: When choosing between conventional irinotecan (e.g., FOLFIRI) and liposomal irinotecan (e.g., nal-IRI/5FU), clinicians must engage in a thoughtful analysis of the available data. In settings where direct comparative trials are lacking (e.g., second-line pancreatic cancer), and retrospective data suggest similar efficacy, the substantially higher cost of the liposomal formulation becomes a critical factor. The decision should be individualized, weighing the potential benefit of reduced cholinergic toxicity with nal-IRI against the significant financial burden it may impose on the patient and the healthcare system.

By integrating these key principles—pharmacogenomic guidance, proactive toxicity management, diligent interaction screening, and value-based decision-making—clinicians can safely harness the potent antitumor activity of irinotecan to maximize its benefit for patients with advanced cancer.

Works cited

  1. Irinotecan - Wikipedia, accessed July 16, 2025, https://en.wikipedia.org/wiki/Irinotecan
  2. Irinotecan - Drugs and Lactation Database (LactMed®) - NCBI Bookshelf, accessed July 16, 2025, https://www.ncbi.nlm.nih.gov/books/NBK500877/
  3. DRUG NAME: Irinotecan - BC Cancer, accessed July 16, 2025, http://www.bccancer.bc.ca/drug-database-site/Drug%20Index/Irinotecan_monograph.pdf
  4. Definition of irinotecan hydrochloride - NCI Drug Dictionary, accessed July 16, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/irinotecan-hydrochloride
  5. Irinotecan | 97682-44-5 - ChemicalBook, accessed July 16, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB1124156.htm
  6. Irinotecan: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed July 16, 2025, https://go.drugbank.com/drugs/DB00762
  7. Individualization of Irinotecan Treatment: A Review of Pharmacokinetics, Pharmacodynamics, and Pharmacogenetics - PubMed, accessed July 16, 2025, https://pubmed.ncbi.nlm.nih.gov/29520731/
  8. Individualization of Irinotecan Treatment: A Review of Pharmacokinetics, Pharmacodynamics, and Pharmacogenetics - PMC, accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6132501/
  9. Irinotecan Free base | CAS#97682-44-5 | AChE inhibitor - MedKoo Biosciences, accessed July 16, 2025, https://www.medkoo.com/products/15034
  10. (+)-Irinotecan | CAS 97682-44-5 | SCBT - Santa Cruz Biotechnology, accessed July 16, 2025, https://www.scbt.com/p/irinotecan-97682-44-5
  11. What is the mechanism of Irinotecan? - Patsnap Synapse, accessed July 16, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-irinotecan
  12. Irinotecan: Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed July 16, 2025, https://www.rxlist.com/irinotecan/generic-drug.htm
  13. FDA approves irinotecan liposome for first-line treatment of ..., accessed July 16, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-irinotecan-liposome-first-line-treatment-metastatic-pancreatic-adenocarcinoma
  14. EMA Approves New First-line Treatment Option for Metastatic Pancreatic Cancer, accessed July 16, 2025, https://digestivecancers.eu/ema-approves-new-first-line-treatment-option-for-metastatic-pancreatic-cancer/
  15. Onivyde pegylated liposomal (previously Onivyde) | European ..., accessed July 16, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/onivyde-pegylated-liposomal
  16. New FDA Approval Brings More Treatment Options in Metastatic Disease - Let's Win Pancreatic Cancer, accessed July 16, 2025, https://letswinpc.org/news/liposomal-irinotecan-approved-pancreatic-cancer/
  17. Liposomal Irinotecan: A Review in Metastatic Pancreatic Adenocarcinoma - PMC, accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7347682/
  18. Second-line Pancreatic Cancer: Nanoliposomal Irinotecan - Targeted Oncology, accessed July 16, 2025, https://www.targetedonc.com/view/second-line-pancreatic-cancer-nanoliposomal-irinotecan
  19. Onivyde® regimen demonstrated statistically significant improvement in overall survival in previously untreated metastatic pancreatic ductal adenocarcinoma - United States - Ipsen, accessed July 16, 2025, https://www.ipsen.com/us/press-releases/onivyde-regimen-demonstrated-statistically-significant-improvement-in-overall-survival-in-previously-untreated-metastatic-pancreatic-ductal-adenocarcinoma/
  20. Onivyde (irinotecan liposomal) FDA Approval History - Drugs.com, accessed July 16, 2025, https://www.drugs.com/history/onivyde.html
  21. CAMPTOSAR® | Pfizer, accessed July 16, 2025, https://www.pfizer.com/products/product-detail/camptosar
  22. Irinotecan Hydrochloride Injection - USA, accessed July 16, 2025, https://www.tevausa.com/our-products/tevagenerics/teva-generics-catalog/vision-product-page/irinotecanhydrochlorideinjection
  23. Irinotecan Pathway, Pharmacokinetics - PharmGKB, accessed July 16, 2025, https://www.pharmgkb.org/pathway/PA2001
  24. The effective combination therapies with irinotecan for colorectal cancer - Frontiers, accessed July 16, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2024.1356708/full
  25. Clinical Pharmacokinetics and Metabolism of Irinotecan (CPT-11) - AACR Journals, accessed July 16, 2025, https://aacrjournals.org/clincancerres/article/7/8/2182/200071/Clinical-Pharmacokinetics-and-Metabolism-of
  26. irinotecan.pdf - Cancer Care Ontario, accessed July 16, 2025, https://www.cancercareontario.ca/en/system/files_force/irinotecan.pdf?download=1
  27. Clinical Pharmacokinetics of Irinotecan and Its Metabolites: A Population Analysis, accessed July 16, 2025, https://ascopubs.org/doi/10.1200/JCO.2002.11.073
  28. Dutch pharmacogenetics working group (DPWG) guideline for the gene–drug interaction between UGT1A1 and irinotecan | Request PDF - ResearchGate, accessed July 16, 2025, https://www.researchgate.net/publication/365801278_Dutch_pharmacogenetics_working_group_DPWG_guideline_for_the_gene-drug_interaction_between_UGT1A1_and_irinotecan
  29. go.drugbank.com, accessed July 16, 2025, https://go.drugbank.com/drugs/DB00762#:~:text=Both%20irinotecan%20and%20SN%2D38,phase%20and%20cancer%20cell%20death.
  30. Irinotecan Pathway, Pharmacodynamics - PharmGKB, accessed July 16, 2025, https://www.pharmgkb.org/pathway/PA2029
  31. Cellular irinotecan resistance in colorectal cancer and overcoming irinotecan refractoriness through various combination trials including DNA methyltransferase inhibitors: a review - ResearchGate, accessed July 16, 2025, https://www.researchgate.net/publication/355850875_Cellular_irinotecan_resistance_in_colorectal_cancer_and_overcoming_irinotecan_refractoriness_through_various_combination_trials_including_DNA_methyltransferase_inhibitors_a_review
  32. Irinotecan|Topoisomerase I inhibitor|CAS# 97682-44-5 - APExBIO, accessed July 16, 2025, https://www.apexbt.com/irinotecan.html
  33. Overcoming Irinotecan Resistance by Targeting Its Downstream Signaling Pathways in Colon Cancer - MDPI, accessed July 16, 2025, https://www.mdpi.com/2072-6694/16/20/3491
  34. Irinotecan: Package Insert / Prescribing Information - Drugs.com, accessed July 16, 2025, https://www.drugs.com/pro/irinotecan.html
  35. www.cancerresearchuk.org, accessed July 16, 2025, https://www.cancerresearchuk.org/about-cancer/treatment/drugs/irinotecan#:~:text=Side%20effects%20during%20or%20soon%20after%20having%20irinotecan&text=The%20symptoms%20include%20diarrhoea%2C%20sweating,irinotecan%20to%20help%20stop%20this.
  36. Irinotecan Monograph for Professionals - Drugs.com, accessed July 16, 2025, https://www.drugs.com/monograph/irinotecan.html
  37. Annotation of FDA Label for irinotecan and UGT1A1 - PharmGKB, accessed July 16, 2025, https://www.pharmgkb.org/labelAnnotation/PA166104831
  38. Managing Irinotecan-Induced Diarrhea: A Comprehensive Review of Therapeutic Interventions in Cancer Treatment - MDPI, accessed July 16, 2025, https://www.mdpi.com/1424-8247/18/3/359/review_report
  39. Clinical pharmacokinetics of irinotecan - PubMed, accessed July 16, 2025, https://pubmed.ncbi.nlm.nih.gov/9342501/
  40. Annotation of DPWG Guideline for irinotecan and UGT1A1, accessed July 16, 2025, https://www.pharmgkb.org/guidelineAnnotation/PA166104951
  41. All You Need to Know About UGT1A1 Genetic Testing for Patients Treated With Irinotecan: A Practitioner-Friendly Guide, accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9014426/
  42. CAMPTOSAR (Irinotecan) Injection, intravenous infusion - accessdata.fda.gov, accessed July 16, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/020571s048lbl.pdf
  43. Clinical Annotation for UGT1A1*1, UGT1A1*28; irinotecan (level 1A Dosage) - PharmGKB, accessed July 16, 2025, https://www.pharmgkb.org/clinicalAnnotation/1451206982
  44. Annotation of EMA Label for irinotecan and UGT1A1 - PharmGKB, accessed July 16, 2025, https://www.pharmgkb.org/labelAnnotation/PA166182952
  45. UGT1A1*28 - PharmGKB, accessed July 16, 2025, https://www.pharmgkb.org/haplotype/PA166115842/prescribingInfo
  46. Dutch pharmacogenetics working group (DPWG) guideline for the gene-drug interaction between UGT1A1 and irinotecan - Maastricht University, accessed July 16, 2025, https://cris.maastrichtuniversity.nl/en/publications/dutch-pharmacogenetics-working-group-dpwg-guideline-for-the-gene-
  47. UGT1A1 CPIC guidelines, accessed July 16, 2025, https://cpicpgx.org/gene/ugt1a1/
  48. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for UGT1A1 and Atazanavir Prescribing - PMC - National Institutes of Health (NIH) |, accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4785051/
  49. CPIC® Guideline for Atazanavir and UGT1A1, accessed July 16, 2025, https://cpicpgx.org/guidelines/guideline-for-atazanavir-and-ugt1a1/
  50. Irinotecan - brand name list from Drugs.com, accessed July 16, 2025, https://www.drugs.com/ingredient/irinotecan.html
  51. Irinotecan Dosage Guide + Max Dose, Adjustments - Drugs.com, accessed July 16, 2025, https://www.drugs.com/dosage/irinotecan.html
  52. Label: CAMPTOSAR- irinotecan hydrochloride injection, solution - DailyMed, accessed July 16, 2025, https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=e518dfc6-7e93-4fee-a66c-51e1ab71c056
  53. Cellular irinotecan resistance in colorectal cancer and overcoming ..., accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8992440/
  54. Phase II trial of irinotecan in patients with progressive or rapidly recurrent colorectal cancer, accessed July 16, 2025, https://pubmed.ncbi.nlm.nih.gov/8648367/
  55. Irinotecan Terminated Phase 2 Trials for Colorectal Cancer Treatment | DrugBank Online, accessed July 16, 2025, https://go.drugbank.com/drugs/DB00762/clinical_trials?conditions=DBCOND0028391&phase=2&purpose=treatment&status=terminated
  56. Irinotecan Completed Phase 2 Trials for Colorectal Cancer / Rectal Cancer / Metastatic Cancer / Cancer / Colon Cancer / Metastatic Colorectal Cancer (CRC) Treatment | DrugBank Online, accessed July 16, 2025, https://go.drugbank.com/drugs/DB00762/clinical_trials?conditions=DBCOND0020966%2CDBCOND0029029%2CDBCOND0028391%2CDBCOND0028461%2CDBCOND0134580%2CDBCOND0029030&phase=2&purpose=treatment&status=completed
  57. Irinotecan Completed Phase 3 Trials for Colorectal Cancer / Metastatic Cancer Treatment, accessed July 16, 2025, https://go.drugbank.com/drugs/DB00762/clinical_trials?conditions=DBCOND0028391%2CDBCOND0028461&phase=3&purpose=treatment&status=completed
  58. Intermittent or Continuous Panitumumab Plus Fluorouracil, Leucovorin, and Irinotecan for First-Line Treatment of RAS and BRAF Wild-Type Metastatic Colorectal Cancer: The IMPROVE Trial | Journal of Clinical Oncology - ASCO Publications, accessed July 16, 2025, https://ascopubs.org/doi/10.1200/JCO.24.00979
  59. FDA Approves Irinotecan Liposome to Treat Pancreatic Cancer - NCI, accessed July 16, 2025, https://www.cancer.gov/news-events/cancer-currents-blog/2015/irinotecan-liposome-pancreatic
  60. Liposomal irinotecan in metastatic pancreatic adenocarcinoma in Asian patients: Subgroup analysis of the NAPOLI‐1 study - PMC - PubMed Central, accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7004519/
  61. Study of MM-398 With or Without 5-FU/​LV, Versus 5-FU/​LV in Patients With Metastatic Pancreatic Cancer (NAPOLI-1) - ClinicalTrials.gov, accessed July 16, 2025, https://clinicaltrials.gov/study/NCT01494506
  62. NAPOLI-1 phase 3 study of liposomal irinotecan in metastatic pancreatic cancer: Final overall survival analysis and characteristics of long-term survivors - PubMed, accessed July 16, 2025, https://pubmed.ncbi.nlm.nih.gov/30654298/
  63. (PDF) NAPOLI-1 phase 3 study of liposomal irinotecan in metastatic ..., accessed July 16, 2025, https://www.researchgate.net/publication/330802218_NAPOLI-1_phase_3_study_of_liposomal_irinotecan_in_metastatic_pancreatic_cancer_Final_overall_survival_analysis_and_characteristics_of_long-term_survivors
  64. EMA Recommends Extension of Therapeutic Indications for Irinotecan Hydrochloride Trihydrate | ESMO, accessed July 16, 2025, https://www.esmo.org/oncology-news/ema-recommends-extension-of-therapeutic-indications-for-irinotecan-hydrochloride-trihydrate
  65. FDA Approves Irinotecan Liposome for Metastatic Pancreatic Adenocarcinoma, accessed July 16, 2025, https://www.pharmacytimes.com/view/fda-approves-irinotecan-liposome-for-metastatic-pancreatic-adenocarcinoma
  66. Ipsen's Onivyde® regimen, a potential new standard-of-care first-line therapy in metastatic pancreatic adenocarcinoma, approved by FDA - United States, accessed July 16, 2025, https://www.ipsen.com/us/press-releases/ipsens-onivyde-regimen-a-potential-new-standard-of-care-first-line-therapy-in-metastatic-pancreatic-adenocarcinoma-approved-by-fda/
  67. A trial looking at irinotecan and carboplatin for small cell lung cancer, accessed July 16, 2025, https://www.cancerresearchuk.org/about-cancer/find-a-clinical-trial/a-trial-looking-at-irinotecan-and-carboplatin-for-small-cell-lung-cancer
  68. Irinotecan and Carboplatin in Treating Patients With Newly Diagnosed Extensive-Stage Small Cell Lung Cancer | ClinicalTrials.gov, accessed July 16, 2025, https://clinicaltrials.gov/study/NCT00104793
  69. RESILIENT Part 2: A Randomized, Open-Label Phase III Study of Liposomal Irinotecan Versus Topotecan in Adults With Relapsed Small Cell Lung Cancer - ASCO Publications, accessed July 16, 2025, https://ascopubs.org/doi/10.1200/JCO.23.02110
  70. Clinical Trial: NCT03088813 - My Cancer Genome, accessed July 16, 2025, https://www.mycancergenome.org/content/clinical_trials/NCT03088813/
  71. Results of a Phase 2 Study Evaluating Lurbinectedin and Irinotecan in Relapsed SCLC Patients With Chemotherapy-Free Interval >30 Days - Conference Correspondent, accessed July 16, 2025, https://conference-correspondent.com/highlights/esmo/esmo-2024/results-of-a-phase-2-study-evaluating-lurbinectedin-and-irinotecan-in-relapsed-sclc-patients-with-chemotherapy-free-interval-30-days
  72. Safe, Prolonged Responses Observed with Lurbinectedin Plus Irinotecan in High-Risk Small Cell Lung Cancer - Targeted Oncology, accessed July 16, 2025, https://www.targetedonc.com/view/safe-prolonged-responses-observed-with-lurbinectedin-plus-irinotecan-in-high-risk-small-cell-lung-cancer
  73. Irinotecan Completed Phase 2 Trials for Breast Cancer Treatment | DrugBank Online, accessed July 16, 2025, https://go.drugbank.com/drugs/DB00762/clinical_trials?conditions=DBCOND0028036&phase=2&purpose=treatment&status=completed
  74. Study Details | Irinotecan in Treating Patients With Refractory Metastatic Breast Cancer, accessed July 16, 2025, https://www.clinicaltrials.gov/study/NCT00003351
  75. Study Details | Irinotecan for Advanced and Metastatic Breast ..., accessed July 16, 2025, https://www.clinicaltrials.gov/study/NCT03562390
  76. Phase II Trial of Bevacizumab and Irinotecan in Recurrent Malignant Glioma | Clinical Cancer Research - AACR Journals, accessed July 16, 2025, https://aacrjournals.org/clincancerres/article/13/4/1253/194479/Phase-II-Trial-of-Bevacizumab-and-Irinotecan-in
  77. Cetuximab, bevacizumab, and irinotecan for patients with primary ..., accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2940618/
  78. Study Details | Bevacizumab and Irinotecan or Temozolomide in Treating Patients With Recurrent or Refractory Glioblastoma Multiforme or Gliosarcoma | ClinicalTrials.gov, accessed July 16, 2025, https://www.clinicaltrials.gov/study/NCT00433381
  79. Triple-drug Therapy With Bevacizumab, Irinotecan, and Temozolomide Plus Tumor Treating Fields for Recurrent Glioblastoma: A Retrospective Study - Frontiers, accessed July 16, 2025, https://www.frontiersin.org/journals/neurology/articles/10.3389/fneur.2019.00042/full
  80. Study Details | Irinotecan And Bevacizumab Combined With Re-radiotherapy in Recurrent Glioblastoma | ClinicalTrials.gov, accessed July 16, 2025, https://clinicaltrials.gov/study/NCT05201326
  81. Irinotecan Completed Phase 1 Trials for Advanced Solid Tumors Treatment - DrugBank, accessed July 16, 2025, https://go.drugbank.com/drugs/DB00762/clinical_trials?conditions=DBCOND0030110&phase=1&purpose=treatment&status=completed
  82. Randomized Phase III Trial of Ramucirumab Beyond Progression Plus Irinotecan in Patients With Ramucirumab-Refractory Advanced Gastric Cancer: RINDBeRG Trial - ASCO Publications, accessed July 16, 2025, https://ascopubs.org/doi/10.1200/JCO.24.01119
  83. Randomized Phase III Trial of Ramucirumab Beyond Progression Plus Irinotecan in Patients With Ramucirumab-Refractory Advanced Gastric Cancer: RINDBeRG Trial - PubMed, accessed July 16, 2025, https://pubmed.ncbi.nlm.nih.gov/40408613/
  84. Randomized Phase III Trial of Ramucirumab Beyond Progression Plus Irinotecan in Patients With Ramucirumab-Refractory Advanced Gastric Cancer: RINDBeRG Trial | Request PDF - ResearchGate, accessed July 16, 2025, https://www.researchgate.net/publication/392027981_Randomized_Phase_III_Trial_of_Ramucirumab_Beyond_Progression_Plus_Irinotecan_in_Patients_With_Ramucirumab-Refractory_Advanced_Gastric_Cancer_RINDBeRG_Trial
  85. Evaluation of second-line apatinib plus irinotecan as a treatment for advanced gastric adenocarcinoma or gastroesophageal conjunction adenocarcinoma: a prospective, multicenter phase II trial - PMC - PubMed Central, accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10166633/
  86. Irinotecan Completed Phase 2 Trials for Small Intestine Adenocarcinoma / Recurrent Small Intestine Cancer Treatment | DrugBank Online, accessed July 16, 2025, https://go.drugbank.com/drugs/DB00762/clinical_trials?conditions=DBCOND0028810%2CDBCOND0028821&phase=2&purpose=treatment&status=completed
  87. Irinotecan and Temozolomide in Combination With Existing High Dose Alkylator Based Chemotherapy for Treatment of Patients With Newly Diagnosed Ewing Sarcoma | ClinicalTrials.gov, accessed July 16, 2025, https://clinicaltrials.gov/study/NCT01864109
  88. Full article: Real-World Outcomes Associated with Liposomal Irinotecan Dose Reductions in Metastatic Pancreatic Ductal Adenocarcinoma - Taylor & Francis Online, accessed July 16, 2025, https://www.tandfonline.com/doi/full/10.2217/fon-2020-0902
  89. Comparison of conventional versus liposomal irinotecan in ..., accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7233326/
  90. Comparison of conventional versus liposomal irinotecan in combination with fluorouracil for advanced pancreatic cancer: a single-institution experience - PubMed, accessed July 16, 2025, https://pubmed.ncbi.nlm.nih.gov/31494781/
  91. Real-World Efficacy Data Reveals OS Boost With Liposomal Irinotecan Plus 5-FU for Metastatic PDAC - CancerNetwork, accessed July 16, 2025, https://www.cancernetwork.com/view/real-world-efficacy-data-reveals-os-boost-with-liposomal-irinotecan-plus-5-fu-for-metastatic-pdac
  92. (PDF) A real-world analysis on the efficacy and tolerability of liposomal irinotecan plus 5-fluorouracil and folinic acid in metastatic pancreatic ductal adenocarcinoma in Belgium - ResearchGate, accessed July 16, 2025, https://www.researchgate.net/publication/373218879_A_real-world_analysis_on_the_efficacy_and_tolerability_of_liposomal_irinotecan_plus_5-fluorouracil_and_folinic_acid_in_metastatic_pancreatic_ductal_adenocarcinoma_in_Belgium
  93. Study Details | A Real-world Study of Liposomal Irinotecan (Onivyde ..., accessed July 16, 2025, https://clinicaltrials.gov/study/NCT07026123
  94. This label may not be the latest approved by FDA. For current ..., accessed July 16, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/020571Orig1s056lbl.pdf
  95. Safety evaluation of irinotecan: a real-world disproportionality analysis using FAERS and JADER databases during the time period 2004-2024 - PubMed Central, accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12184384/
  96. CAMPTO® - (Irinotecan hydrochloride) - Pfizer, accessed July 16, 2025, https://labeling.pfizer.com/ShowLabeling.aspx?id=15268
  97. DEP® irinotecan phase 2 commences after positive phase 1 results - Starpharma, accessed July 16, 2025, https://starpharma.com/news/view/485/dep-irinotecan-phase-2-commences-after-positive-phase-1-results
  98. Onivyde - This label may not be the latest approved by FDA. For current labeling information, please visit https://www.fda.gov/drugsatfda, accessed July 16, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/207793s016lbl.pdf
  99. Camptosar irinotecan hydrochloride injection For Intravenous Use Only WARNINGS CAMPTOSAR Injection should be administered only u, accessed July 16, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/020571s031s032s033s036s037lbl.pdf
  100. Camptosar® (Irinotecan) - GlobalRPH, accessed July 16, 2025, https://globalrph.com/oncology/camptosar-irinotecan/
  101. Chemotherapy-Induced Diarrhea: Options for Treatment and Prevention, accessed July 16, 2025, https://jhoponline.com/issue-archive/2012-issues/december-2012-vol-3-no-4/15408:chemotherapy-unduced-diarrhea-options
  102. Herbal Medicines for Irinotecan-Induced Diarrhea - Frontiers, accessed July 16, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2019.00182/full
  103. Irinotecan Injection: MedlinePlus Drug Information, accessed July 16, 2025, https://medlineplus.gov/druginfo/meds/a608043.html
  104. Irinotecan (intravenous route) - Side effects & uses - Mayo Clinic, accessed July 16, 2025, https://www.mayoclinic.org/drugs-supplements/irinotecan-intravenous-route/description/drg-20064383
  105. Managing Irinotecan-Induced Diarrhea: A Comprehensive Review ..., accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11944746/
  106. Irinotecan-Induced Diarrhea during a Protracted Administration Schedule for Pediatric Sarcomas - Mechanisms and Clinical Applica, accessed July 16, 2025, https://www.clinicsinoncology.com/open-access/irinotecan-induced-diarrhea-during-a-protractednbspadministration-schedule-for-pediatric-9390.pdf
  107. Camptosar (irinotecan) dosing, indications, interactions, adverse effects, and more, accessed July 16, 2025, https://reference.medscape.com/drug/camptosar-irinotecan-342252
  108. Irinotecan Intravenous: Uses, Side Effects, Interactions, Pictures, Warnings & Dosing, accessed July 16, 2025, https://www.webmd.com/drugs/2/drug-13700/irinotecan-intravenous/details
  109. For the Patient: Irinotecan - BC Cancer, accessed July 16, 2025, http://www.bccancer.bc.ca/drug-database-site/Drug%20Index/Irinotecan_handout.pdf
  110. Irinotecan Interactions Checker - Drugs.com, accessed July 16, 2025, https://www.drugs.com/drug-interactions/irinotecan.html
  111. Irinotecan liposomal Interactions - Drugs.com, accessed July 16, 2025, https://www.drugs.com/drug-interactions/irinotecan-liposomal.html
  112. Irinotecan Disease Interactions - Drugs.com, accessed July 16, 2025, https://www.drugs.com/disease-interactions/irinotecan.html
  113. Irinotecan Actavis - NEW ZEALAND DATA SHEET, accessed July 16, 2025, https://www.medsafe.govt.nz/profs/datasheet/i/irinotecanactavisinj.pdf
  114. Evolution of resistance to Irinotecan in cancer cells involves generation of topoisomerase-guided mutations in non-coding genome that reduce the chances of DNA breaks | bioRxiv, accessed July 16, 2025, https://www.biorxiv.org/content/10.1101/2021.11.26.470089.full
  115. Evolution of resistance to Irinotecan in cancer cells involves generation of topoisomerase-guided mutations in non-coding genome that reduce the chances of DNA breaks | bioRxiv, accessed July 16, 2025, https://www.biorxiv.org/content/10.1101/2021.11.26.470089v3
  116. Mechanisms and Strategies to Overcome Drug Resistance in Colorectal Cancer - MDPI, accessed July 16, 2025, https://www.mdpi.com/1422-0067/26/5/1988
  117. Strategic Combinations to Prevent and Overcome Resistance to Targeted Therapies in Oncology - ASCO Publications, accessed July 16, 2025, https://ascopubs.org/doi/10.1200/EDBK_280845
  118. Phase II study of IRInotecan treatment after COmbined chemo-immunotherapy for extensive-stage small cell lung cancer: Protocol of IRICO study - PubMed, accessed July 16, 2025, https://pubmed.ncbi.nlm.nih.gov/37675546/
  119. Phase II study of IRInotecan treatment after COmbined chemo ..., accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10542459/
  120. Pharmacokinetic Study of Lurbinectedin in Combination With Irinotecan in Patients With Selected Solid Tumors | ClinicalTrials.gov, accessed July 16, 2025, https://www.clinicaltrials.gov/study/NCT02611024
  121. Novel Camptothecin Derivatives - In Vivo, accessed July 16, 2025, https://iv.iiarjournals.org/content/invivo/19/1/283.full-text.pdf

Published at: July 16, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.