SN-38: A Comprehensive Monograph on a Pivotal Topoisomerase I Inhibitor—From Prodrug Metabolite to Advanced Therapeutic Payload
1.0 Introduction: The Re-emergence of a Potent Camptothecin Analog
The history of oncology is marked by the discovery of natural products with profound cytotoxic activity, whose clinical potential was initially hindered by challenging pharmaceutical properties. Few molecules exemplify this narrative better than SN-38 (7-ethyl-10-hydroxycamptothecin). As a semi-synthetic analog of camptothecin, SN-38 belongs to a class of agents that revolutionized cancer treatment by identifying a novel molecular target: DNA topoisomerase I.[1] However, SN-38 is most widely known not as a drug administered to patients, but as the principal active metabolite of irinotecan (CPT-11), a cornerstone chemotherapeutic agent used in the treatment of various solid tumors, most notably metastatic colorectal cancer.[3] The entire therapeutic rationale for administering the water-soluble prodrug irinotecan is to facilitate the
in vivo generation of SN-38, the molecule responsible for virtually all of the desired antitumor effect.[6]
This prodrug strategy was born from a central paradox that defined the early development of SN-38. On one hand, its cytotoxic potency is immense; in vitro assays consistently demonstrate that SN-38 is 100 to 2,000 times more active than irinotecan itself, making it one of the most powerful topoisomerase I inhibitors known.[4] On the other hand, this potency was historically shackled by profoundly unfavorable physicochemical characteristics. SN-38 is extremely hydrophobic, rendering it virtually insoluble in water and other pharmaceutically acceptable solvents.[9] Furthermore, its chemically essential lactone ring is unstable at physiological pH, rapidly hydrolyzing to a pharmacologically inert carboxylate form.[9] These properties made direct intravenous formulation and administration of SN-38 a formidable challenge, effectively preventing its development as a standalone clinical agent for decades.
The story of SN-38 is therefore a compelling case study in the evolution of pharmaceutical sciences, illustrating a paradigm shift from systemic prodrug strategies toward highly engineered, targeted therapeutics. The inherent limitations of SN-38, coupled with the inefficient and highly variable enzymatic conversion from irinotecan that leads to unpredictable patient toxicity and efficacy, served as a direct catalyst for innovation.[14] This "pharmaceutical challenge" drove researchers to devise novel methods to solubilize, stabilize, and deliver SN-38 directly to tumor tissues. This has led to a modern renaissance for the molecule, re-emerging from the shadow of its prodrug to become a critical payload in a new generation of advanced drug delivery systems. These include its encapsulation within nanocarriers like liposomes and polymeric micelles, and, most notably, its conjugation to monoclonal antibodies to form antibody-drug conjugates (ADCs).[10] The landmark approval of Sacituzumab Govitecan (Trodelvy), an ADC that uses SN-38 as its cytotoxic payload, for the treatment of metastatic breast cancer and other solid tumors, represents the culmination of these efforts and has firmly re-established SN-38 at the forefront of cancer therapy.[18]
This monograph provides a definitive and integrated analysis of SN-38. It traces the molecule's journey from its fundamental chemical properties and pharmacological mechanism through its complex pharmacokinetics as a metabolite, the critical role of pharmacogenomics in its toxicity, and its ultimate rebirth as a key component of rationally designed, next-generation cancer therapeutics.
2.0 Molecular Profile and Physicochemical Characteristics
A comprehensive understanding of SN-38 begins with its precise molecular identity and the physicochemical properties that have so profoundly influenced its development and clinical application.
2.1 Identification and Nomenclature
To establish a definitive reference point, SN-38 is identified by a range of systematic names and registry numbers across chemical and pharmacological databases.
- Primary Name: SN-38 [3]
- Systematic (IUPAC) Name: (4S)-4,11-diethyl-4,9-dihydroxy-1,4-dihydro-3H,14H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinoline-3,14-dione [4]
- Common Synonyms: 7-Ethyl-10-hydroxycamptothecin, 7-ethyl-10-hydroxy-20(S)-Camptothecin, SN 38 lactone, NK 012.[3] It is important to note that synonyms such as "Camptosar," "CPT-11," and "irinotecan" refer specifically to the prodrug and are incorrectly applied to SN-38, though this conflation occasionally appears in the literature.[3]
- Registry Numbers:
- CAS Number: 86639-52-3 [3]
- DrugBank ID: DB05482 [24]
- PubChem CID: 104842 [3]
- Other Identifiers: ChEBI: 8988; ChEMBL: ChEMBL837; UNII: 0H43101T0J [4]
2.2 Chemical Structure and Properties
SN-38 is classified as a small molecule, plant-derived quinoline alkaloid.[16] Its structure and properties are fundamental to its biological activity.
- Chemical Formula: C22H20N2O5 [3]
- Molecular Weight: The molecular weight is consistently reported as approximately 392.4 g/mol or Da.[3]
- Structural Features: The molecule possesses a rigid, planar pentacyclic (5-ring) structure. The most critical feature for its anticancer activity is the α-hydroxy-lactone ring (Ring E) at the 20(S) chiral center. The integrity of this lactone ring is absolutely essential for binding to its molecular target, topoisomerase I.[26] The planar nature of the fused ring system allows the molecule to intercalate into the DNA helix at the site of enzyme action.
- Cheminformatics Data: For computational modeling and database searching, the following identifiers are used:
- Canonical SMILES: CCC1=C2CN3C(=O)C4=C(COC(=O)C4(CC)O)C=C3C2=NC5=C1C=C(C=C5)O [4]
- InChI Key: FJHBVJOVLFPMQE-QFIPXVFZSA-N [4]
- Computed Properties: Analysis of its structure predicts several key properties relevant to its behavior as a drug. It has 5 hydrogen bond acceptors, 2 hydrogen bond donors, and 2 rotatable bonds. Its topological polar surface area (TPSA) is calculated to be 101.65 A˚2.[28] These values provide insight into its limited solubility and its ability to permeate cell membranes, a key feature for its "bystander effect" in ADCs.
Property | Value | Source(s) |
---|
CAS Number | 86639-52-3 | 3 |
DrugBank ID | DB05482 | 24 |
PubChem CID | 104842 | 3 |
Molecular Formula | C22H20N2O5 | 3 |
Molecular Weight | ~392.4 g/mol | 3 |
IUPAC Name | (4S)-4,11-diethyl-4,9-dihydroxy-1,4-dihydro-3H,14H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinoline-3,14-dione | 4 |
Appearance | Crystalline solid; white to yellow to green powder | 16 |
Melting Point | ~217 °C | 16 |
Solubility (Water) | Insoluble / Poor / Sparingly soluble | 10 |
Solubility (Organic) | DMSO: ~2-50 mg/mL; Ethanol: Insoluble | 21 |
Key Structural Feature | Pentacyclic alkaloid with essential α-hydroxy-lactone ring | 26 |
Table 2.1: Key Identifiers and Physicochemical Properties of SN-38. This table consolidates the most critical data points for SN-38, providing a validated summary for researchers and highlighting the foundational challenge of its poor water solubility. | | |
2.3 Solubility and Stability: The Core Challenge
The clinical development of SN-38 has been dominated by efforts to overcome two fundamental physicochemical flaws: its poor solubility and its instability in physiological conditions.
- Aqueous Solubility: SN-38 is consistently described as being "extremely hydrophobic," "virtually insoluble in water," and "sparingly soluble in aqueous buffers".[9] This property makes it impossible to formulate as a simple aqueous solution for intravenous administration, which was the primary obstacle to its direct clinical use.[12]
- Organic Solvent Solubility: For in vitro laboratory research and formulation development, SN-38 is soluble in polar aprotic organic solvents like dimethyl sulfoxide (DMSO), with reported solubilities ranging from 2 mg/mL to as high as 50 mg/mL with warming and sonication.[21] It is also soluble in dimethylformamide (DMF) but is largely insoluble in other common solvents like ethanol.[21]
- Lactone Ring Instability: Beyond solubility, the chemical stability of SN-38 is a major liability. The active form of the drug contains a six-membered lactone ring (Ring E), which is prone to hydrolysis. At neutral or alkaline pH (specifically, pH > 6.0), the lactone ring reversibly opens to form a pharmacologically inactive carboxylate species.[9] Because normal physiological pH is ~7.4, a significant portion of the drug rapidly converts to this inactive form upon entering the bloodstream, drastically reducing its therapeutic potential. This inactive carboxylate form also binds with high affinity to human serum albumin, which further sequesters it away from its intended target in tumor cells.[13] Strategies to overcome these issues, such as encapsulation in nanocarriers or complexation with cyclodextrins, are designed specifically to protect the lactone ring from hydrolysis and improve solubility.[10]
2.4 Safety and Handling Specifications
As a potent cytotoxic agent, SN-38 requires careful handling in laboratory and manufacturing environments.
- Physical State: It is supplied as a crystalline solid, with an appearance that can range from white to yellow or even green powder.[16]
- Hazard Classification: SN-38 is classified as a toxic and hazardous substance. The signal word is "Danger," and it carries multiple hazard statements, including H301 (Toxic if swallowed), H315 (Causes skin irritation), H319 (Causes serious eye irritation), and H335 (May cause respiratory irritation).[33] Appropriate personal protective equipment is mandatory when handling the compound.
- Transportation and Storage: For shipping, it is classified under UN numbers UN1544 or UN2811 (Alkaloids, solid, n.o.s. or Toxic Solid, Organic, N.O.S.) with Hazard Class 6.1 and Packing Group III.[16] The compound is heat-sensitive and must be stored at low temperatures, typically frozen at <0°C or -20°C, to ensure long-term stability.[16]
3.0 Pharmacology: Mechanism of Cytotoxic Action
The potent antineoplastic activity of SN-38 stems from its specific and highly effective disruption of a fundamental cellular process: DNA replication and maintenance. Its mechanism is centered on the poisoning of the nuclear enzyme DNA topoisomerase I.
3.1 Primary Target: DNA Topoisomerase I
The sole molecular target of SN-38 and other camptothecin analogs is DNA Topoisomerase I (Topo I).[5] Topo I is an essential enzyme present in all eukaryotic cells. Its primary physiological function is to resolve topological problems that arise in the DNA double helix during critical processes like replication, transcription, and repair. It accomplishes this by inducing transient, reversible single-strand breaks (SSBs) in the DNA backbone, which allows the DNA to unwind and relieve torsional strain. The enzyme then rapidly re-ligates the break to restore the integrity of the DNA strand.[5] This process is vital for preventing the accumulation of lethal DNA tangles and breaks.
3.2 The Ternary "Cleavable Complex" and DNA Damage Cascade
SN-38 does not inhibit Topo I in the classical sense of blocking its active site. Instead, it functions as an "interfacial poison." Its mechanism proceeds through the following steps:
- Stabilization of the Cleavable Complex: SN-38 exerts its effect by binding to the transient intermediate state where Topo I is covalently linked to the 3'-phosphate end of the broken DNA strand. The planar, pentacyclic structure of SN-38 allows it to intercalate into the DNA at this cleavage site, stacking against the flanking base pairs.[8] This interaction stabilizes the so-called "cleavable complex," physically preventing the enzyme from completing its catalytic cycle and re-ligating the DNA strand.[5]
- Conversion to Double-Strand Breaks: This trapping of the cleavable complex transforms a fleeting, harmless SSB into a persistent DNA lesion. The ultimate cytotoxic event occurs when a moving DNA replication fork, advancing along the DNA template during S-phase, collides with this stabilized ternary complex (DNA-Topo I-SN-38). This collision leads to the collapse of the replication fork and converts the single-strand break into a highly toxic, irreversible double-strand break (DSB).[5]
This mechanism explains why SN-38 is predominantly an S-phase-specific agent. Its lethality is maximized in cells that are actively replicating their DNA. This S-phase specificity is a classic "double-edged sword." It is the very reason SN-38 is so effective against rapidly proliferating cancer cells, which have a large fraction of their population in S-phase at any given time. However, it is also the direct mechanistic cause of the drug's most significant dose-limiting toxicities, which occur in the body's normal tissues with high cell turnover rates, namely the bone marrow (leading to myelosuppression and neutropenia) and the gastrointestinal epithelium (leading to mucositis and diarrhea).[4] The drug's efficacy and its primary side effects are thus inextricably linked through its core mechanism of action.
While the S-phase-dependent model is dominant, some evidence suggests a more nuanced mechanism. Studies have shown that SN-38 can induce chromatid breaks immediately following exposure, independent of DNA synthesis. The chromosomal aberrations observed immediately after treatment are qualitatively different from those seen 24 hours later, which are clearly associated with DNA replication (e.g., radial chromosome configurations).[35] This indicates that while the most potent cytotoxic pathway is S-phase dependent, SN-38 may also exert a degree of clastogenic (chromosome-damaging) activity through a secondary, S-phase-independent mechanism.
3.3 Cellular Consequences and Apoptosis Induction
The generation of lethal DSBs by SN-38 triggers a cascade of cellular responses culminating in programmed cell death, or apoptosis.
- Cell Cycle Arrest: The presence of extensive DNA damage activates cellular checkpoint pathways, which halt the cell cycle to allow time for repair. SN-38 exposure leads to a robust arrest of cells in the S and G2 phases, preventing them from entering mitosis with damaged chromosomes.[7]
- Apoptotic Cascade: When the DNA damage is too severe to be repaired, the cell initiates the intrinsic apoptotic pathway. Key events in this process induced by SN-38 include:
- Collapse of Mitochondrial Membrane Potential (MMP): A critical early and irreversible step in apoptosis, leading to the release of pro-apoptotic factors like cytochrome c from the mitochondria.[38]
- Caspase Activation: The released factors trigger the activation of a cascade of cysteine proteases known as caspases. SN-38 robustly activates the key executioner enzyme, caspase-3.[38]
- PARP Cleavage: Activated caspase-3 proceeds to cleave numerous cellular substrates, including Poly (ADP-ribose) polymerase (PARP). The cleavage and inactivation of PARP is a classic hallmark of apoptosis and serves to prevent futile DNA repair attempts, conserving cellular energy for the process of cell dismantling.[38]
- Inhibition of Macromolecular Synthesis: As a direct consequence of its interference with DNA topology, SN-38 is a potent inhibitor of both DNA and RNA synthesis. Reported half-maximal inhibitory concentrations (IC50) for these processes are approximately 0.077 µM for DNA synthesis and 1.3 µM for RNA synthesis.[23]
3.4 Potency and Comparative Cytotoxicity
A defining characteristic of SN-38 is its extraordinary potency. It is consistently reported to be orders of magnitude more cytotoxic than its parent prodrug, irinotecan. While the exact ratio varies depending on the cell line and assay conditions, a general consensus places SN-38 as being 100- to 1,000-fold more potent than irinotecan, with some reports extending this range from 2- to 2,000-fold.[4]
In vitro studies confirm its activity at very low concentrations. For example, in lung cancer cell lines, cytotoxic effects are observed at just 10 nM, with an IC50 around 100 nM.[38] In the HT-29 colon cancer cell line, the IC50 is even lower, at a potent 8.8 nM.[29] This high intrinsic potency is the fundamental reason it is such a desirable payload for targeted delivery systems.
4.0 Clinical Pharmacokinetics and Metabolism
The clinical behavior of SN-38 is inseparable from that of its prodrug, irinotecan. The complex, multi-step pathway from irinotecan administration to SN-38 clearance is a major determinant of both the drug's efficacy and its toxicity, and it underpins the critical role of pharmacogenomics in its safe use.
4.1 The Irinotecan-SN-38 Axis: An Inefficient Prodrug System
Irinotecan (CPT-11) was developed as a water-soluble prodrug specifically to overcome the formulation challenges of camptothecin and its analogs, including SN-38.[3] The therapeutic activity of irinotecan is almost entirely dependent on its metabolic conversion to SN-38.
- Activation Pathway: This activation occurs primarily in the liver and other tissues containing carboxylesterase enzymes (CES), such as CES1 and CES2. These enzymes hydrolyze the bulky bipiperidino side chain of irinotecan to yield the active metabolite, SN-38.[4]
- Inefficiency and Variability: A crucial limitation of this system is its inefficiency. The CES enzymes have a relatively low affinity for irinotecan, resulting in a very small fraction of the administered dose—typically less than 10%, and often reported to be as low as <3%—being converted to the active SN-38 form.[14] Furthermore, the activity of CES enzymes varies significantly among individuals, contributing to the large interpatient variability observed in SN-38 exposure, clinical response, and toxicity.[8]
4.2 Absorption, Distribution, Metabolism, and Excretion (ADME) of SN-38
Once formed from irinotecan, SN-38 follows its own distinct pharmacokinetic pathway.
- Distribution: SN-38 exhibits a high degree of binding to plasma proteins, particularly albumin. Approximately 95% of SN-38 in the circulation is protein-bound.[8] This is substantially higher than the protein binding of irinotecan (~65%) and means that only a small fraction of the total SN-38 concentration is free and pharmacologically active.[8] It has a large volume of distribution, indicating extensive partitioning into tissues.[8]
- Metabolism (Detoxification): The primary pathway for the inactivation and detoxification of SN-38 is glucuronidation. This metabolic reaction occurs predominantly in the liver, where the enzyme UDP-glucuronosyltransferase 1A1 (UGT1A1) conjugates a glucuronic acid moiety to the 10-hydroxy group of SN-38.[4] This process converts the lipophilic, active SN-38 into a more water-soluble and virtually inactive metabolite, SN-38 glucuronide (SN-38G).[14] This single enzyme, UGT1A1, is responsible for the vast majority of SN-38 clearance.
- Excretion and Enterohepatic Recirculation: Irinotecan and its metabolites, including SN-38 and SN-38G, are primarily eliminated from the body via the biliary route, with subsequent excretion in the feces.[8] This excretory pathway is the source of one of the drug's most problematic toxicities. The inactive SN-38G, having been excreted into the intestinal lumen, can be targeted by β-glucuronidase enzymes produced by the resident gut microflora. These bacterial enzymes cleave the glucuronide conjugate, reversing the detoxification process and liberating the active, highly toxic SN-38 directly within the gut.[14] This locally re-activated SN-38 can then exert its potent cytotoxic effects on the rapidly dividing cells of the intestinal mucosa. This process of biliary excretion and subsequent intestinal re-activation is a form of enterohepatic recirculation and is a major contributor to the severe, delayed-onset diarrhea associated with irinotecan therapy.[26] The entire pharmacokinetic journey of irinotecan and SN-38 effectively creates a "perfect storm" for gastrointestinal toxicity, combining systemic exposure with a mechanism for localized, high-concentration re-exposure within the sensitive gut environment.
- Pharmacokinetic Parameters: Systemic plasma concentrations of SN-38 are approximately 100-fold lower than those of the parent irinotecan.[8] Following a standard intravenous infusion of irinotecan, peak plasma concentrations (Cmax) of SN-38 are typically reached after about one hour.[8] The plasma decay of SN-38 closely follows that of irinotecan, with a long apparent terminal half-life ranging from 6 to 30 hours.[8] Critically, the area under the plasma concentration-time curve (AUC) for SN-38—a measure of total drug exposure—increases proportionally with the administered dose of irinotecan and is significantly correlated with the incidence and severity of both neutropenia and diarrhea.[8]
The profound reliance on a single, highly polymorphic enzyme (UGT1A1) for the clearance of the primary toxic moiety (SN-38) makes this drug a poster child for the field of pharmacogenomics. The metabolic fate of the drug, and consequently the safety of the patient, is heavily dependent on an individual's genetic makeup. An inability to efficiently clear SN-38 due to genetic factors leads directly to higher and more prolonged exposure to the active drug, which in turn drives the risk of severe, life-threatening toxicity. This makes UGT1A1 genotyping an essential tool for personalizing therapy and ensuring patient safety.
5.0 Clinical Application, Toxicity, and Pharmacogenomic Considerations
The clinical use of SN-38 is defined by the profile of its prodrug, irinotecan, and is characterized by a narrow therapeutic window where efficacy is balanced against significant, predictable toxicities. Mitigating these toxicities through pharmacogenomic-guided dosing has become a central aspect of its clinical management.
5.1 Clinical Development and Investigational Use
Because SN-38 is the active metabolite of irinotecan, it is implicitly used in all of irinotecan's approved indications. These include a primary role in the treatment of metastatic colorectal cancer, often as part of combination regimens like FOLFIRI (fluorouracil, leucovorin, irinotecan), as well as in pancreatic cancer, lung cancer, and other solid tumors.[3]
Efforts to administer SN-38 directly, bypassing the inefficient irinotecan prodrug step, have focused on advanced formulations. A notable example is liposome-encapsulated SN-38 (LE-SN38), which was evaluated in a Phase 1 clinical trial (NCT00046540) for patients with advanced solid tumors.[43] Other investigational trials have explored SN-38 for specific indications, including a Phase 2 trial in small cell lung cancer (NCT00104754, subsequently withdrawn) and a currently recruiting Phase 1 trial in head and neck cancer (NCT04640480).[48] In a sign of its recognized potential, the European Medicines Agency (EMA) granted orphan drug designation in 2019 to a 7-ethyl-10-hydroxy-camptothecin formulation for the treatment of soft tissue sarcoma.[3]
5.2 The Toxicity Profile: A Direct Consequence of SN-38 Exposure
The dose-limiting toxicities (DLTs) associated with irinotecan therapy are not caused by the prodrug itself but are directly attributable to the cytotoxic effects of its active metabolite, SN-38, on healthy, rapidly-dividing tissues.[4]
- Severe Diarrhea: This is the most infamous and often most severe toxicity, affecting a substantial portion of patients and potentially leading to dehydration, electrolyte imbalances, and hospitalization.[4] It manifests in two distinct forms:
- Early-onset diarrhea: Occurring within the first 24 hours of infusion, this form is caused by a cholinergic effect (inhibition of acetylcholinesterase) and can typically be managed or prevented with atropine.[26]
- Late-onset diarrhea: Occurring more than 24 hours after administration, this form is more severe, prolonged, and unpredictable. It is caused by the direct cytotoxic damage to the intestinal mucosa from SN-38 that has been excreted into the gut via the bile and subsequently re-activated by bacterial enzymes, as described in Section 4.2.[5]
- Myelosuppression (Neutropenia): The other major DLT is severe (Grade 3-4) neutropenia, the suppression of neutrophil production in the bone marrow.[5] The risk and severity of neutropenia are directly correlated with the plasma AUC of SN-38.[8] This can lead to the serious complication of febrile neutropenia, where a low neutrophil count is accompanied by fever, increasing the risk of life-threatening infections.[41]
- Other Common Adverse Events: In addition to the DLTs, patients frequently experience alopecia (hair loss), nausea, vomiting, and fatigue, all of which are characteristic side effects of cytotoxic chemotherapy and are linked to the SN-38 payload.[20]
5.3 The Role of UGT1A1 Polymorphisms: The Key to Personalized Dosing
The observation that toxicity varies dramatically between patients led to the discovery of a critical genetic determinant: polymorphisms in the UGT1A1 gene. As this enzyme is solely responsible for detoxifying SN-38, its functional status is a key predictor of a patient's risk for severe adverse events.[4]
- The UGT1A1*28 Allele: The most common and well-studied polymorphism is the *28 allele (also known as UGT1A1(TA)7). The promoter region of the wild-type UGT1A1 gene contains six tandem repeats of the sequence "TA" ((TA)6TAA). The *28 allele contains seven repeats ((TA)7TAA).[18] This extra repeat in the TATA box region of the promoter impairs gene transcription, leading to significantly reduced expression of the UGT1A1 enzyme and, consequently, a decreased capacity to glucuronidate and clear SN-38.[4]
- Clinical Risk by Genotype:
- **Homozygous Wild-Type (e.g., 1/1 or 6/6): These patients have normal enzyme activity and are at standard risk for toxicity.
- **Heterozygous (e.g., 1/28 or 6/7): These patients have one normal and one variant allele, resulting in intermediate enzyme activity and a moderately increased risk of toxicity.
- **Homozygous Variant (e.g., 28/28 or 7/7): These patients have two copies of the variant allele, leading to markedly reduced enzyme activity. They are at the highest risk for developing severe, life-threatening neutropenia and diarrhea when treated with standard doses of irinotecan.[18] Many individuals with the benign hereditary condition Gilbert's syndrome, characterized by mild hyperbilirubinemia, are homozygous for the *28 allele.[45]
- Other Alleles of Concern: While *28 is the most common variant in Caucasian and African populations, other functional polymorphisms exist. For example, the *6 allele (a missense mutation, Gly71Arg) also results in reduced enzyme activity and is particularly prevalent in East Asian populations, where it confers a similar high risk of toxicity.[18]
Allele | Genetic Change | Functional Effect on Enzyme | Population Prevalence (Approx.) | Clinical Implication for Homozygotes |
---|
UGT1A1*1 | (TA)6TAA promoter | Normal expression and activity | N/A (Wild Type) | Standard risk of toxicity. |
UGT1A1*28 | (TA)7TAA promoter | Reduced gene transcription and enzyme expression | 40% in European/African ancestry | High risk of severe neutropenia and diarrhea. Reduced starting dose of irinotecan is recommended. |
UGT1A1*6 | c.211G>A (Gly71Arg) | Reduced catalytic activity | 15% in East Asian ancestry | High risk of severe neutropenia. Reduced starting dose is recommended. |
UGT1A1*93 | c.-3156G>A | Reduced gene transcription | 27-34% in European/African ancestry | Associated with increased SN-38 exposure and risk of neutropenia. |
Table 5.2: Common UGT1A1 Polymorphisms and Their Clinical Impact on SN-38 Metabolism and Toxicity. This table translates complex genetic data into a clinically actionable format, providing the evidence-based rationale for UGT1A1 genotyping as a critical component of personalized medicine when using SN-38-releasing agents. Data synthesized from.4 | | | | |
5.4 Regulatory Guidance and Dosing Implications
The strong, well-established link between UGT1A1 genotype and SN-38-mediated toxicity prompted regulatory action. In 2005, the U.S. Food and Drug Administration (FDA) revised the drug label for irinotecan (Camptosar®) to include information about this pharmacogenomic association. The label now explicitly warns of the increased risk of severe neutropenia in patients homozygous for the UGT1A1*28 allele and recommends considering a reduced initial dose for these individuals.[18] This has paved the way for the clinical implementation of pre-emptive pharmacogenomic testing. By identifying high-risk patients before the first dose, clinicians can proactively adjust the dose to improve the safety and tolerability of treatment.[18] This principle was further validated in the Phase 1 trial of LE-SN38, which prospectively stratified patients by genotype and confirmed that *28/*28 patients had 2- to 3-fold greater drug exposure than wild-type patients at the same dose level, underscoring the necessity of genotype-guided dosing for any SN-38-based therapy.[43]
6.0 Overcoming Limitations: Advanced Drug Delivery Systems
The clinical trajectory of SN-38 has been fundamentally reshaped by innovations in drug delivery. These advanced platforms were rationally designed to systematically address each of the molecule's inherent flaws, transforming it from a "difficult" metabolite into a highly effective therapeutic payload.
6.1 Rationale for Novel Formulations
The development of sophisticated delivery systems for SN-38 was driven by a clear set of objectives aimed at solving its core challenges:
- Enhance Solubility: To overcome its extreme hydrophobicity and enable intravenous administration.[9]
- Protect the Lactone Ring: To shield the active form of the drug from hydrolysis at physiological pH, thereby preventing its inactivation in the bloodstream.[9]
- Bypass Inefficient Prodrug Conversion: To deliver SN-38 directly to the body, eliminating the reliance on the inefficient and highly variable CES-mediated conversion from irinotecan and its associated interpatient variability.[15]
- Improve the Therapeutic Index: To enhance tumor-specific delivery through active or passive targeting, thereby concentrating the cytotoxic agent at the site of disease and reducing systemic exposure to healthy tissues.[10]
6.2 Nanoparticle-Based Formulations: Encapsulating Potency
One major strategy has been to encapsulate SN-38 within various types of nanoparticles, which serve as protective transport vehicles.
- Liposomal SN-38 (LE-SN38): This was a pioneering effort in SN-38 delivery. Liposomes are microscopic, spherical vesicles composed of a lipid bilayer that can encapsulate hydrophobic drugs like SN-38 within their core.[39] The Phase 1 trial of LE-SN38 (NCT00046540) yielded critical findings: the formulation was generally well-tolerated, and, most importantly, it did not cause the severe, late-onset diarrhea that is the hallmark toxicity of conventional irinotecan.[43] This strongly suggested that the liposomal encapsulation successfully altered the drug's biodistribution, preventing high concentrations of free SN-38 from accumulating in the gut. The trial also reinforced the importance of UGT1A1 genotyping for determining the appropriate dose.[43]
- Polymeric Micelles and Prodrugs: This approach involves chemically conjugating SN-38 to amphiphilic polymers, which then self-assemble in aqueous solution to form nanoscopic micelles with a drug-containing core and a hydrophilic shell.[10] A common technique is PEGylation, the attachment of polyethylene glycol (PEG) chains. PEG-SN-38 conjugates can act as long-circulating macromolecular prodrugs that slowly release SN-38 over time. This strategy aims to maintain a sustained therapeutic concentration of the drug while avoiding the high peak concentrations (Cmax) that are often associated with acute toxicity.[15]
- Other Emerging Nanocarriers: The field continues to evolve with the investigation of other innovative platforms:
- Cubosomes: These are advanced liquid crystalline nanoparticles with a unique cubic internal structure that can encapsulate SN-38 with very high efficiency and provide excellent stability for the lactone ring.[32]
- Core-Shell Nanoparticles: These are complex, multi-component systems, such as those with an oxaliplatin-based core and a lipid shell containing a cholesterol-conjugated SN-38 prodrug. Such platforms are being designed for the co-delivery of multiple chemotherapeutic agents and may offer synergistic effects, particularly in the context of chemo-immunotherapy.[54]
- Protein Nanoparticles: Natural proteins like human serum albumin are also being explored as biocompatible nanocarriers for SN-38.[10]
6.3 SN-38 as an ADC Payload: The Sacituzumab Govitecan (Trodelvy) Case Study
The most clinically successful and transformative application of SN-38 delivery technology to date is its use as the cytotoxic payload in the antibody-drug conjugate (ADC) Sacituzumab Govitecan (SG, brand name Trodelvy®).
- Components and Rational Design: SG is a highly engineered therapeutic consisting of three distinct components designed to work in synergy [20]:
- Antibody: Sacituzumab (hRS7), a humanized monoclonal antibody that specifically targets Trophoblast cell-surface antigen-2 (Trop-2). Trop-2 is a transmembrane protein that is highly overexpressed on the surface of a wide variety of epithelial tumors—including breast, urothelial, lung, and others—but has limited expression on most normal, healthy tissues.[13]
- Payload: SN-38, the potent topoisomerase I inhibitor, chosen for its extreme cytotoxicity.[18]
- Linker: A proprietary, hydrolyzable linker called CL2A. This linker covalently attaches SN-38 to the antibody via a carbonate bond. It is engineered to be stable in the bloodstream during circulation (pH 7.4) but is susceptible to hydrolysis and cleavage in the acidic environment of the tumor cell's endosomes and lysosomes (pH < 6.0).[13] SG features a relatively high drug-to-antibody ratio (DAR) of approximately 7.6, meaning each antibody carries multiple molecules of the payload.[37]
- Targeted Mechanism of Action: The therapeutic action of SG follows a precise, multi-step process:
- SG circulates in the bloodstream and binds with high affinity to Trop-2 on the surface of cancer cells.
- Upon binding, the entire ADC-Trop-2 complex is rapidly internalized by the cell via endocytosis.
- The complex is trafficked intracellularly to lysosomes.
- The acidic environment within the lysosome cleaves the hydrolyzable CL2A linker, releasing the free, unmodified SN-38 payload directly into the cytoplasm of the cancer cell.
- The released SN-38 is then free to diffuse to the nucleus, where it inhibits topoisomerase I and induces the apoptotic cascade as previously described.[37]
- The Bystander Effect: A Key Advantage for Overcoming Heterogeneity: A crucial feature that distinguishes SG and contributes significantly to its efficacy is the "bystander effect." This phenomenon is a direct consequence of the rational selection of both the payload and the linker. Because the released SN-38 payload is a small, relatively lipophilic, and uncharged molecule, it is membrane-permeable. This allows it to diffuse out of the originally targeted Trop-2-positive cell and enter adjacent tumor cells, killing them regardless of whether they express the Trop-2 antigen.[13] This is a powerful mechanism for overcoming tumor heterogeneity, a common cause of treatment failure where not all cancer cells within a tumor express the target antigen uniformly. The bystander effect effectively allows the ADC to function as a tool for localized, high-concentration chemotherapy delivery, expanding its cytotoxic reach throughout the tumor microenvironment.
- Clinical Success: The intelligent design of SG has translated into remarkable clinical success. Based on the results of pivotal clinical trials, such as the ASCENT study, which demonstrated dramatic improvements in progression-free survival and overall survival compared to standard single-agent chemotherapy, SG has received FDA and EMA approval for the treatment of heavily pretreated metastatic triple-negative breast cancer (mTNBC) and, more recently, for hormone receptor-positive (HR+)/HER2-negative metastatic breast cancer.[18] Its toxicity profile, featuring neutropenia and diarrhea, is a direct reflection of the SN-38 payload and is managed with supportive care and dose modifications, with UGT1A1*28 status remaining a significant risk factor for increased toxicity.[46]
7.0 Comparative Analysis and Future Perspectives
The enduring relevance of SN-38 in oncology is best understood by comparing it to other agents in its class and by examining the ongoing research aimed at further refining its therapeutic application.
7.1 SN-38 vs. Other Topoisomerase I Inhibitors
While SN-38 (delivered via irinotecan or ADCs) is a leading Topo I inhibitor, it is not the only one in clinical use.
- Comparison with Topotecan: Topotecan is the other major clinically approved camptothecin analog and serves as an important benchmark.[2] The two drugs, despite sharing the same molecular target, have distinct clinical profiles driven by their different pharmacokinetic properties.
- Metabolism and Clearance: A key difference lies in their administration and elimination. Topotecan is administered as an active drug and is primarily cleared by the kidneys, meaning dose adjustments may be necessary in patients with renal impairment. In contrast, irinotecan is a prodrug that requires hepatic activation to SN-38, and SN-38 is then cleared by the liver via UGT1A1-mediated glucuronidation. This makes hepatic function and UGT1A1 genetics the critical factors for irinotecan/SN-38 dosing.[51]
- Potency: In vitro studies comparing the two active drugs have generally found SN-38 to be significantly more potent than topotecan in cell lines derived from hematological and ovarian cancers.[66]
- Clinical Niche and Toxicity: These differences translate to distinct clinical roles and toxicity profiles. Irinotecan/SN-38 is a cornerstone of therapy for gastrointestinal cancers, particularly colorectal cancer, and its DLTs are famously diarrhea and neutropenia. Topotecan is more established in the treatment of ovarian cancer and small-cell lung cancer (SCLC), with its DLTs being primarily hematologic (neutropenia and thrombocytopenia).[19] Recent head-to-head trials of liposomal irinotecan versus topotecan in relapsed SCLC found that while overall survival was similar, the irinotecan formulation produced a much higher objective response rate but also had its own distinct safety profile.[70]
Characteristic | SN-38 (delivered via Irinotecan) | Topotecan |
---|
Administration Form | Prodrug (Irinotecan) | Active Drug |
Primary Activation | Hepatic (Carboxylesterases) | N/A |
Primary Clearance | Hepatic (UGT1A1 glucuronidation) | Renal |
Key Dose-Limiting Toxicities | Diarrhea, Neutropenia | Neutropenia, Thrombocytopenia, Anemia |
Primary Clinical Use | Colorectal Cancer, Pancreatic Cancer | Ovarian Cancer, Small Cell Lung Cancer |
Intrinsic Potency | Very High | High |
Table 7.1: Comparative Profile of SN-38 (via Irinotecan) and Topotecan. This table provides a concise summary of the key differences between the two main clinical topoisomerase I inhibitors, highlighting how their divergent pharmacokinetic pathways lead to distinct clinical applications and toxicity profiles. Data synthesized from.2 | | |
- Comparison with Next-Generation Payloads (e.g., Exatecan/DXd): The field of ADC payloads is continuously evolving. Newer Topo I inhibitors, such as exatecan and its derivative DXd (the payload in Trastuzumab Deruxtecan, or Enhertu®), have been developed. DXd is reported to be approximately 10 times more potent than SN-38 and also possesses a strong bystander effect.[27] This demonstrates the ongoing search for even more powerful payloads and represents the next step in the evolution of this drug class.
7.2 Mechanisms of Resistance
As with all effective anticancer agents, tumors can develop resistance to SN-38, limiting its long-term efficacy. The primary mechanisms of resistance include [26]:
- Target Alteration: The most direct mechanism is a change in the drug's target, Topo I. This can involve the downregulation of Topo I protein expression, reducing the number of available targets, or mutations in the TOP1 gene that alter the drug's binding site, preventing it from effectively stabilizing the cleavable complex.[26]
- Enhanced Drug Efflux: Cancer cells can increase their expression of ATP-binding cassette (ABC) membrane transporters, which function as drug efflux pumps. The transporter ABCG2 (also known as Breast Cancer Resistance Protein, BCRP) has been identified as a key mediator of SN-38 resistance, actively pumping the drug out of the cell and reducing its intracellular concentration.[34]
- Activation of Pro-Survival Pathways: Resistant cells can compensate for the drug's cytotoxic effects by upregulating signaling pathways that promote cell survival and proliferation. This can include the activation of the Epidermal Growth Factor Receptor (EGFR) pathway or the MAPK p38 survival pathway.[34]
7.3 Emerging Therapeutic Strategies
Research into SN-38 is now focused on leveraging its potency in novel therapeutic combinations and delivery strategies to overcome resistance and further improve outcomes.
- Combination with Immunotherapy: A highly promising area of investigation is the combination of SN-38-based therapies with immune checkpoint inhibitors (e.g., anti-PD-1/PD-L1 antibodies). Preclinical evidence suggests a powerful synergy. SN-38 may enhance the effects of immunotherapy by modulating the tumor microenvironment; studies have shown it can decrease the expression of the immunosuppressive ligand PD-L1 on tumor cells while simultaneously increasing the infiltration and cytotoxic activity of antitumor immune cells, such as natural killer (NK) cells and CD8+ T-cells.[54] This combination of direct cytotoxic killing and immune system activation represents a potent strategy for achieving more durable responses.
- Novel Prodrug and Linker Technologies: The success of Sacituzumab Govitecan has spurred further research into even more sophisticated delivery systems. This includes the design of novel linkers and prodrugs that are responsive to specific stimuli within the tumor microenvironment. For example, systems are being developed with linkers that are cleaved in response to high levels of reactive oxygen species (ROS) or glutathione (GSH), which are often elevated in tumors. Light-sensitive systems are also being explored. These strategies aim to achieve "on-demand" drug release with even greater tumor specificity, further widening the therapeutic window of SN-38.[6]
8.0 Conclusion: The Enduring Relevance of SN-38 in Oncology
The journey of SN-38 is a remarkable testament to the persistence and ingenuity of pharmaceutical science. Initially identified as the highly potent but pharmaceutically challenging active metabolite of irinotecan, it spent decades as a "problem child" of drug development, its immense potential locked away by its poor solubility and instability. The very difficulties it presented, however, became the impetus for innovation, driving the creation of novel solutions to deliver this powerful agent effectively and safely.
Through a deep, integrated understanding of its molecular structure, pharmacological mechanism, complex pharmacokinetics, and the genetic basis of its toxicity, researchers have successfully "tamed" SN-38. The evolution from an inefficient systemic prodrug system to precisely engineered nanocarriers and antibody-drug conjugates represents a microcosm of the broader shift in oncology towards precision medicine. The clinical success of Sacituzumab Govitecan has validated SN-38 as a premier ADC payload, demonstrating that its inherent flaws can be completely overcome through rational drug design. This platform technology not only solves the historical formulation issues but also adds new layers of efficacy, such as the bystander effect, to combat the clinical challenge of tumor heterogeneity.
Today, SN-38 stands not as a failed drug candidate, but as a cornerstone of modern targeted therapy. Its future lies not in its use as a standalone agent, but in its continued and expanding role as the cytotoxic warhead in ever-more sophisticated delivery platforms. As research into novel combinations, such as chemo-immunotherapy, and next-generation stimuli-responsive systems continues, the enduring relevance of SN-38 in the therapeutic armamentarium against cancer is firmly secured, cementing its legacy as a molecule that has been successfully and brilliantly repurposed for the era of precision oncology.
Works cited
- Definition of camptothecin analog - NCI Dictionary of Cancer Terms, accessed August 1, 2025, https://www.cancer.gov/publications/dictionaries/cancer-terms/def/camptothecin-analog
- Comparison of In Vitro Activities of Camptothecin and Nitidine Derivatives against Fungal and Cancer Cells | Antimicrobial Agents and Chemotherapy - ASM Journals, accessed August 1, 2025, https://journals.asm.org/doi/10.1128/aac.43.12.2862
- 7-Ethyl-10-Hydroxycamptothecin | C22H20N2O5 | CID 104842 - PubChem, accessed August 1, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/7-Ethyl-10-hydroxycamptothecin
- SN-38 - Wikipedia, accessed August 1, 2025, https://en.wikipedia.org/wiki/SN-38
- What is the mechanism of Irinotecan Hydrochloride? - Patsnap Synapse, accessed August 1, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-irinotecan-hydrochloride
- Irinotecan and its active metabolite, SN-38: Review of bioanalytical methods and recent update from clinical pharmacology perspectives | Request PDF - ResearchGate, accessed August 1, 2025, https://www.researchgate.net/publication/38031567_Irinotecan_and_its_active_metabolite_SN-38_Review_of_bioanalytical_methods_and_recent_update_from_clinical_pharmacology_perspectives
- Irinotecan: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed August 1, 2025, https://go.drugbank.com/drugs/DB00762
- Clinical pharmacokinetics of irinotecan - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/9342501/
- SN-38: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed August 1, 2025, https://go.drugbank.com/drugs/DB05482
- SN38 Drug Delivery System in Cancer Treatment | IJN, accessed August 1, 2025, https://www.dovepress.com/research-progress-of-sn38-drug-delivery-system-in-cancer-treatment-peer-reviewed-fulltext-article-IJN
- SN-38-Cyclodextrin Complexation and Its Influence on the Solubility, Stability, and In Vitro Anticancer Activity Against Ovarian Cancer - PMC, accessed August 1, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3969487/
- Phase solubility diagram of SN-38 in water (pH adjusted to 3 with 0.02... - ResearchGate, accessed August 1, 2025, https://www.researchgate.net/figure/Phase-solubility-diagram-of-SN-38-in-water-pH-adjusted-to-3-with-002-N-HCl-at-room_fig3_259985725
- The Mode of Action and Clinical Outcomes of Sacituzumab Govitecan in Solid Tumors, accessed August 1, 2025, https://aacrjournals.org/clincancerres/article/31/8/1390/754522/The-Mode-of-Action-and-Clinical-Outcomes-of
- Irinotecan Pathway, Pharmacokinetics - PharmGKB, accessed August 1, 2025, https://www.pharmgkb.org/pathway/PA2001
- Macromolecular Prodrug That Provides the Irinotecan (CPT-11) Active-Metabolite SN-38 with Ultralong Half-Life, Low Cmax , and Low Glucuronide Formation - ACS Publications, accessed August 1, 2025, https://pubs.acs.org/doi/10.1021/jm401644v
- 7-Ethyl-10-hydroxycamptothecin 86639-52-3 - TCI Chemicals, accessed August 1, 2025, https://www.tcichemicals.com/US/en/p/E0748
- SN-38 - BPS Bioscience, accessed August 1, 2025, https://bpsbioscience.com/sn-38-82257
- All You Need to Know About UGT1A1 Genetic Testing for Patients ..., accessed August 1, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9014426/
- Camptothein-Based Anti-Cancer Therapies and Strategies to ... - MDPI, accessed August 1, 2025, https://www.mdpi.com/2072-6694/17/6/1032
- Sacituzumab Govitecan for Metastatic Triple‐Negative Breast Cancer: Clinical Overview and Management of Potential Toxicities - PMC - PubMed Central, accessed August 1, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8488774/
- SN-38 (CAS 86639-52-3) - Cayman Chemical, accessed August 1, 2025, https://www.caymanchem.com/product/15632/sn-38
- 7-Ethyl-10-hydroxycamptothecin | CAS 86639-52-3 | TCI-E0748 | Spectrum Chemical, accessed August 1, 2025, https://www.spectrumchemical.com/7-ethyl-10-hydroxycamptothecin-tci-e0748
- SN-38 (7-Ethyl-10-hydroxycamptothecin) | Topoisomerase I Inhibitor | MedChemExpress, accessed August 1, 2025, https://www.medchemexpress.com/SN-38.html
- DGIdb, accessed August 1, 2025, https://www.dgidb.org/drugs/drugbank:DB05482
- www.tocris.com, accessed August 1, 2025, https://www.tocris.com/products/sn-38_2684#:~:text=Preparing%20Stock%20Solutions%20for%20SN,the%20product%20molecular%20weight%20392.4.
- TOPOISOMERASE I – TARGETING DRUGS - PRINCIPLES OF ANTINEOPLASTIC THERAPY, accessed August 1, 2025, https://hemonc.medicine.ufl.edu/files/2013/07/TopoI.pdf
- Topoisomerase I inhibitors Based ADCs: SN-38 & DXd | Biopharma PEG, accessed August 1, 2025, https://www.biochempeg.com/article/376.html
- SN-38 | Ligand page - IUPHAR/BPS Guide to PHARMACOLOGY, accessed August 1, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?tab=structure&ligandId=6925
- cdn.caymanchem.com, accessed August 1, 2025, https://cdn.caymanchem.com/cdn/insert/15632.pdf
- SN 38 | DNA topoisomerase Inhibitor | Buy from Supplier AdooQ®, accessed August 1, 2025, https://www.adooq.com/sn-38.html
- SN-38 | 99.96%(HPLC) | In Stock | ADC Cytotoxin inhibitor - Selleckchem.com, accessed August 1, 2025, https://www.selleckchem.com/products/sn-38.html
- Preparation and Characterization of SN-38-Encapsulated Phytantriol Cubosomes Containing α-Monoglyceride Additives - J-Stage, accessed August 1, 2025, https://www.jstage.jst.go.jp/article/cpb/64/6/64_c15-00984/_html/-char/en
- 7-Ethyl-10-hydroxycamptothecin - Oakwood Chemical, accessed August 1, 2025, https://www.oakwoodchemical.com/ProductsList.aspx?CategoryID=-2&txtSearch=40245&ExtHyperLink=1
- Irinotecan Pathway, Pharmacodynamics - PharmGKB, accessed August 1, 2025, https://www.pharmgkb.org/pathway/PA2029
- Topoisomerase-I inhibitor SN-38 can induce DNA damage and chromosomal aberrations independent from DNA synthesis - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/9858931/
- Resistance to TOP-1 Inhibitors: Good Old Drugs Still Can Surprise Us - MDPI, accessed August 1, 2025, https://www.mdpi.com/1422-0067/24/8/7233
- Trodelvy® Mechanism of Action - Gilead Medical Information, accessed August 1, 2025, https://www.askgileadmedical.com/docs/trodelvy/trodelvy-mechanism-of-action/pdf
- Topoisomerase I inhibitor SN-38 effectively attenuates growth of ..., accessed August 1, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3133571/
- Preclinical Safety, Pharmacokinetics and Antitumor Efficacy Profile of Liposome-entrapped SN-38 Formulation - Anticancer Research, accessed August 1, 2025, https://ar.iiarjournals.org/content/anticanres/25/1A/331.full-text.pdf
- Structure of irinotecan and its active metabolite SN-38 - ResearchGate, accessed August 1, 2025, https://www.researchgate.net/figure/Figure-1-Structure-of-irinotecan-and-its-active-metabolite-SN-38_fig1_341035219
- Clinical Pharmacology Biopharmaceutics Review(s) - accessdata.fda.gov, accessed August 1, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2004/20-571s021_Camptosar_biopharmr.pdf
- Physiologically based pharmacokinetic modeling for predicting irinotecan exposure in human body | Oncotarget, accessed August 1, 2025, https://www.oncotarget.com/article/18380/text/
- Final results of a phase I study of liposome encapsulated SN-38 (LE-SN38): Safety, pharmacogenomics, pharmacokinetics, and tumor response - ASCO Publications, accessed August 1, 2025, https://ascopubs.org/doi/10.1200/jco.2005.23.16_suppl.2017
- Population pharmacokinetics and pharmacodynamics of irinotecan (CPT-11) and active metabolite SN-38 during phase I trials - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/7786822/
- UGT1A1 polymorphism predicts irinotecan toxicity: Evolving proof, accessed August 1, 2025, https://www.periodicos.capes.gov.br/index.php/acervo/buscador.html?task=detalhes&id=W2042793526
- Trodelvy, INN-sacituzumab govitecan - EMA, accessed August 1, 2025, https://www.ema.europa.eu/en/documents/product-information/trodelvy-epar-product-information_en.pdf
- SN-38 Completed Phase 1 Trials for Neoplasms Treatment | DrugBank Online, accessed August 1, 2025, https://go.drugbank.com/drugs/DB05482/clinical_trials?conditions=DBCOND0017273&phase=1&purpose=treatment&status=completed
- Head And Neck Cancer Recruiting Phase 1 Trials for SN-38 (DB05482) | DrugBank Online, accessed August 1, 2025, https://go.drugbank.com/indications/DBCOND0042625/clinical_trials/DB05482?phase=1&status=recruiting
- SN-38 Withdrawn Phase 2 Trials for Lung Cancer Treatment | DrugBank Online, accessed August 1, 2025, https://go.drugbank.com/drugs/DB05482/clinical_trials?conditions=DBCOND0028388&phase=2&purpose=treatment&status=withdrawn
- Model-Based Prediction of Nanoparticle and Dissolved Form Ratios Using Total Concentration Data: A Case Study of SNB-101 - Frontiers, accessed August 1, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2025.1556618/full
- Overview of Irinotecan and Topotecan: The Topoisomerase I ..., accessed August 1, 2025, https://rxpharmacist.com/overview-of-irinotecan-and-topotecan-the-topoisomerase-i-inhibitors/
- The role of SN-38 exposure, UGT1A1*28 polymorphism, and baseline bilirubin level in predicting severe irinotecan toxicity - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/17192505/
- Pharmacogenomic and pharmacokinetic assessment of liposome encapsulated SN-38 (LE-SN38) in advanced cancer patients - ASCO Publications, accessed August 1, 2025, https://ascopubs.org/doi/10.1200/jco.2004.22.90140.2501
- Two-Stage SN38 Release from a Core–Shell Nanoparticle Enhances Tumor Deposition and Antitumor Efficacy for Synergistic Combination with Immune Checkpoint Blockade | ACS Nano, accessed August 1, 2025, https://pubs.acs.org/doi/10.1021/acsnano.2c09788
- What is the mechanism of action of Sacituzumab tirumotecan? - Patsnap Synapse, accessed August 1, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-action-of-sacituzumab-tirumotecan
- A Closer Look at Sacituzumab Govitecan-hziy - Hematology & Oncology, accessed August 1, 2025, https://www.hematologyandoncology.net/archives/november-2020/a-closer-look-at-sacituzumab-govitecan-hziy/
- Mechanism of Action (MOA) TRODELVY® (sacituzumab govitecan-hziy) | Official HCP Site, accessed August 1, 2025, https://www.trodelvyhcp.com/moa
- Sacituzumab govitecan: Uses, Interactions, Mechanism of Action, accessed August 1, 2025, https://go.drugbank.com/drugs/DB12893
- go.drugbank.com, accessed August 1, 2025, https://go.drugbank.com/drugs/DB12893#:~:text=Sacituzumab%20govitecan%20is%20sacituzumab%20govitecan,induce%20DNA%20damage%2Dmediated%20apoptosis.
- www.askgileadmedical.com, accessed August 1, 2025, https://www.askgileadmedical.com/docs/trodelvy/trodelvy-mechanism-of-action/pdf#:~:text=Pharmacology%20data%20suggest%20that%20SG,to%20apoptosis%20and%20cell%20death.
- The Bystander Effect of ADCs | Biopharma PEG, accessed August 1, 2025, https://www.biochempeg.com/article/269.html
- Trodelvy® (sacituzumab govitecan-hziy) Mechanism of Action - Gilead Medical Information, accessed August 1, 2025, https://www.askgileadmedical.com/docs/trodelvy/trodelvy-mechanism-of-action
- Antibody–Drug Conjugate Sacituzumab Govitecan Drives Efficient Tissue Penetration and Rapid Intracellular Drug Release | Molecular Cancer Therapeutics - AACR Journals, accessed August 1, 2025, https://aacrjournals.org/mct/article/22/1/102/711995/Antibody-Drug-Conjugate-Sacituzumab-Govitecan
- Sacituzumab govitecan: past, present and future of a new antibody-drug conjugate and future horizon - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/36069628/
- Camptothecin (CPT) and its derivatives are known to target topoisomerase I (Top1) as their mechanism of action: did we miss something in CPT analogue molecular targets for treating human disease such as cancer? - PMC - PubMed Central, accessed August 1, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5752681/
- pubmed.ncbi.nlm.nih.gov, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/11044651/#:~:text=In%20haematological%20and%20ovarian%20cancer,with%20clinical%20experiences%20to%20date.
- Jonsson E, Dhar S, Jonsson B, Nygren P, Graf W, Larsson RDifferential activity of topotecan, irinotecan and SN-38 in fresh human tumour cells but not in cell lines. Eur J Cancer 36: 2120-2127 - ResearchGate, accessed August 1, 2025, https://www.researchgate.net/publication/12279147_Jonsson_E_Dhar_S_Jonsson_B_Nygren_P_Graf_W_Larsson_RDifferential_activity_of_topotecan_irinotecan_and_SN-38_in_fresh_human_tumour_cells_but_not_in_cell_lines_Eur_J_Cancer_36_2120-2127
- Differential activity of topotecan, irinotecan and SN-38 in fresh human tumour cells but not in cell lines - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/11044651/
- Camptothecin and Its Derivatives from Traditional Chinese Medicine in Combination with Anticancer Therapy Regimens: A Systematic Review and Meta-Analysis - MDPI, accessed August 1, 2025, https://www.mdpi.com/2072-6694/16/22/3802
- www.targetedonc.com, accessed August 1, 2025, https://www.targetedonc.com/view/liposomal-irinotecan-shows-comparable-efficacy-with-topotecan-in-relapsed-sclc#:~:text=Liposomal%20Irinotecan%20Shows%20Comparable%20Efficacy%20With%20Topotecan%20in%20Relapsed%20SCLC,-Jun%2018%2C%202024&text=While%20the%20primary%20end%20point,relapsed%20small%20cell%20lung%20cancer.
- Liposomal Irinotecan vs Topotecan in Relapsed Small Cell Lung Cancer, accessed August 1, 2025, https://ascopost.com/news/may-2024/liposomal-irinotecan-vs-topotecan-in-relapsed-small-cell-lung-cancer/
- Liposomal Irinotecan Shows Comparable Efficacy With Topotecan in Relapsed SCLC, accessed August 1, 2025, https://www.targetedonc.com/view/liposomal-irinotecan-shows-comparable-efficacy-with-topotecan-in-relapsed-sclc
- Irinotecan Liposomal Injection Shows No OS Improvement vs Topotecan in SCLC, accessed August 1, 2025, https://www.targetedonc.com/view/irinotecan-liposomal-injection-shows-no-os-improvement-vs-topotecan-in-sclc
- Molecular characterization of irinotecan (SN-38) resistant human breast cancer cell lines - RePub, Erasmus University Repository, accessed August 1, 2025, https://repub.eur.nl/pub/90564/REPUB_90564-OA.pdf
- SN-38, an active metabolite of irinotecan, enhances anti-PD-1 treatment efficacy in head and neck squamous cell carcinoma - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/36641765/