Capecitabine (DB01101): A Comprehensive Monograph on its Pharmacology, Clinical Efficacy, and Therapeutic Landscape
Executive Summary
Capecitabine is an orally administered fluoropyrimidine carbamate that has become a cornerstone chemotherapeutic agent in the modern oncology armamentarium.[1] Identified by DrugBank ID DB01101 and CAS Number 154361-50-9, it functions as a rationally designed, systemic prodrug that is relatively inert until it undergoes a multi-step enzymatic conversion to its active cytotoxic moiety, 5-fluorouracil (5-FU).[1] A key feature of its design is a tumor-selective activation process, driven by the higher concentrations of the enzyme thymidine phosphorylase in many malignant tissues compared to healthy ones. This mechanism is intended to generate high local concentrations of 5-FU at the site of malignancy, thereby enhancing the therapeutic index and mimicking the pharmacokinetic profile of a continuous 5-FU infusion without the associated complexities of intravenous administration.[1]
The clinical utility of capecitabine is well-established, with regulatory approvals from the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for a broad spectrum of solid tumors. Its primary indications include the adjuvant and metastatic treatment of colorectal cancer, the management of advanced or metastatic breast cancer, and integral roles in combination regimens for gastric, esophageal, and pancreatic cancers.[1]
The safety profile of capecitabine is significant but generally manageable with appropriate monitoring and dose adjustments. The most common dose-limiting toxicities include palmar-plantar erythrodysesthesia (hand-foot syndrome) and diarrhea.[7] A critical safety concern, highlighted by a boxed warning, is the potential for a severe drug interaction with vitamin K antagonists like warfarin, which can lead to life-threatening bleeding.[4] Furthermore, its use is contraindicated in patients with a known complete deficiency of the enzyme dihydropyrimidine dehydrogenase (DPD), a critical pharmacogenomic consideration that can lead to severe toxicity.[7]
Capecitabine represents a landmark in oral chemotherapy, offering patients significant convenience and an established efficacy profile. Its clinical application continues to evolve through extensive research into novel combination therapies, particularly with targeted agents and immunotherapies, and the exploration of optimized dosing schedules designed to mitigate toxicity while preserving efficacy.[9] This ongoing investigation ensures its enduring relevance in a rapidly advancing therapeutic landscape.
Compound Profile: Physicochemical and Structural Characterization
A precise understanding of capecitabine's chemical and physical properties is fundamental to appreciating its formulation, stability, and biological behavior. This section provides a comprehensive characterization of the molecule, consolidating data from chemical databases, regulatory filings, and commercial suppliers.
Nomenclature and Identifiers
To ensure unambiguous identification across scientific literature, clinical practice, and regulatory documentation, capecitabine is defined by a standardized set of names and identifiers.
- Generic Name: Capecitabine [1]
- Brand Names: The originator product is marketed as Xeloda®.[1] Numerous generic versions and other brand names are available globally, including Ecansya, Xitabin, and Kapetral.[1]
- Systematic (IUPAC) Name: pentyl N--5-fluoro-2-oxopyrimidin-4-yl]carbamate.[5]
- CAS Number: 154361-50-9.[12]
- Database Identifiers:
- DrugBank ID: DB01101 [1]
- PubChem CID: 60953 [2]
- ChemSpider ID: 54916 [2]
- ChEBI ID: 31348 [2]
- NSC Code: 712807 [21]
- Internal Research Code: Ro 09-1978 [12]
Molecular and Structural Formulae
The atomic composition and spatial arrangement of capecitabine are defined by its molecular and structural formulas.
- Molecular Formula: C15H22FN3O6. This formula is consistently reported across authoritative sources.[6] An erroneous formula of
C13H24EN20 appears in some literature, which is inconsistent with the compound's structure and likely represents a typographical error in the source publication.[23]
- Molecular Weight: The calculated molecular weights are highly consistent:
- Average Mass: 359.35 g/mol [4]
- Monoisotopic Mass: 359.149264 Da [12]
- Chemical Structure Representations: For computational chemistry and database cross-referencing, the structure is represented by:
- SMILES: CCCCCOC(=O)NC1=NC(=O)N(C=C1F)[C@H]2[C@@H]([C@@H]([C@H](O2)C)O)O [6]
- InChIKey: GAGWJHPBXLXJQN-UORFTKCHSA-N [6]
Physicochemical Properties
The physical characteristics of capecitabine dictate its formulation, handling, and behavior in biological systems.
- Appearance: Capecitabine is a white to off-white or beige crystalline solid or powder.[4]
- Solubility: The solubility profile is critical for its oral formulation and absorption.
- Aqueous: The definitive aqueous solubility is stated as 26 mg/mL at 20°C in the FDA label [4], a value corroborated by other sources.[15] Reports of lower solubility likely reflect different conditions, such as pH, as evidenced by a value of 0.15 mg/mL in PBS at pH 7.2.[14] The characterization as "not in water" in some commercial literature is a practical oversimplification.[12]
- Organic Solvents: The compound is freely soluble in ethanol (e.g., 207 mg/mL) and DMSO (e.g., 72 mg/mL), and soluble in DMF (e.g., 14 mg/mL).[14]
- Stability and Storage: As a solid, capecitabine is stable for at least four years.[14] Recommended storage conditions are typically refrigerated at 2-8°C [17] or frozen at -20°C for long-term storage.[12] It is stable enough for shipment at ambient temperature for short durations.[12] Solutions are less stable and should ideally be prepared fresh; however, they can be stored for up to one month at -20°C.[15]
- Hazard Information: Capecitabine is classified as a hazardous and potentially dangerous drug that requires special handling and disposal procedures.[7] It carries hazard classifications of Carcinogenicity Category 1B, Mutagenicity Category 2, and Reproductive Toxicity Category 1B.[17]
The drug's specific physicochemical properties are not merely descriptive details; they are the essential attributes that permit its existence as a successful oral chemotherapeutic. Its adequate aqueous solubility and high stability as a crystalline solid are the physical basis for its formulation into a reliable, orally administered tablet. This formulation provides a profound clinical advantage over intravenous 5-FU, fundamentally improving patient quality of life by reducing time spent in infusion centers. The classification as a hazardous drug carries direct and important implications for the safety of pharmacists, nurses, patients, and caregivers, mandating specific protocols for handling and administration to prevent inadvertent exposure.[7]
Property | Value | Source(s) |
---|
Chemical Class | Fluoropyrimidine Carbamate; Antimetabolite | 1 |
CAS Number | 154361-50-9 | 14 |
Molecular Formula | C15H22FN3O6 | 14 |
Molecular Weight | 359.35 g/mol (Average) | 4 |
Appearance | White to off-white crystalline powder | 4 |
Aqueous Solubility | 26 mg/mL (at 20°C) | 4 |
Organic Solubility | Soluble in DMSO; Freely soluble in Ethanol | 15 |
Storage (Solid) | 2-8°C or -20°C (long term) | 12 |
Stability (Solid) | ≥ 4 years | 14 |
Hazard Classification | Carc. 1B, Muta. 2, Repr. 1B | 17 |
Table 1: Key Chemical and Physical Properties of Capecitabine | | |
Pharmacodynamics: The Molecular Basis of Antineoplastic Activity
The clinical efficacy of capecitabine is rooted in its sophisticated pharmacodynamic profile as a rationally designed prodrug. It belongs to the fluoropyrimidine carbamate class of drugs and functions as a nucleoside metabolic inhibitor and an antimetabolite.[1] In its parent form, capecitabine is relatively non-cytotoxic and must undergo a precise, multi-step bioactivation process to generate its ultimate effector molecule, 5-fluorouracil (5-FU), which then disrupts fundamental cellular processes in malignant cells.[4]
The Bioactivation Cascade: A Three-Step Enzymatic Conversion
The journey from inert prodrug to active chemotherapeutic is a sequential, three-step enzymatic cascade that occurs in different tissues, a pathway that is central to its pharmacological strategy.
- Step 1: Hepatic Hydrolysis. Following oral administration and absorption, capecitabine first travels to the liver. Here, it is extensively hydrolyzed by the enzyme carboxylesterase to an intermediate metabolite, 5'-deoxy-5-fluorocytidine (5'-DFCR).[4] This initial step begins the unmasking of the drug's cytotoxic potential.
- Step 2: Deamination. The 5'-DFCR metabolite is subsequently converted to a second intermediate, 5'-deoxy-5-fluorouridine (5'-DFUR), also known as doxifluridine.[14] This reaction is catalyzed by the enzyme cytidine deaminase, which is found in high concentrations in the liver but is also present in various tumor tissues.[4]
- Step 3: Tumor-Localized Activation. The final and most critical step in the bioactivation pathway is the conversion of 5'-DFUR into the active cytotoxic agent, 5-fluorouracil (5-FU). This conversion is catalyzed by the enzyme thymidine phosphorylase (dThdPase), also known as platelet-derived endothelial cell growth factor.[4]
Tumor-Selective Activation
The therapeutic advantage of capecitabine over systemically administered 5-FU is derived from this final activation step. The design of capecitabine was predicated on the observation that many human cancers, including breast, colorectal, and gastric tumors, express significantly higher levels of thymidine phosphorylase (dThdPase) compared to surrounding healthy tissues.[1] This differential enzyme expression allows for the preferential conversion of 5'-DFUR to 5-FU within the tumor microenvironment. The result is a high local concentration of the active drug at the desired site of action, which serves to maximize antineoplastic activity while simultaneously minimizing systemic exposure and the associated toxicities common with intravenous 5-FU, particularly severe gastrointestinal side effects.[1] This elegant design effectively allows capecitabine to mimic the pharmacokinetics of a continuous 5-FU infusion, but with the convenience and safety advantages of an oral agent.[1] The dependence on dThdPase for activation suggests that its expression level within a tumor could serve as a predictive biomarker for treatment efficacy, a concept supported by preclinical data showing a correlation between dThdPase levels and antitumor activity.[18] While not yet a standard clinical practice, this presents a potential avenue for future patient stratification and personalized therapy.
Cytotoxic Mechanisms of 5-Fluorouracil (5-FU)
Once generated within the tumor, 5-FU exerts its potent anticancer effects through the action of its own active metabolites, which launch a dual-pronged attack on nucleic acid synthesis and function, ultimately leading to cell death.[1]
- Inhibition of DNA Synthesis: A primary mechanism of 5-FU cytotoxicity involves its metabolism to 5-fluoro-2'-deoxyuridine monophosphate (FdUMP).[1] FdUMP bears a strong structural resemblance to the natural substrate deoxyuridylate. It forms a highly stable, covalently bound ternary complex with the enzyme thymidylate synthase (TS) and its essential folate cofactor,
N5,10-methylenetetrahydrofolate. This irreversible binding potently inhibits TS, thereby blocking the methylation of deoxyuridylate to thymidylate. Thymidylate is the rate-limiting precursor for the synthesis of thymidine triphosphate (TTP), an essential building block for DNA replication and repair. The resulting depletion of the TTP pool leads to an imbalance of deoxynucleotides, inhibition of DNA synthesis, and ultimately, cell death, a process often referred to as "thymineless death".[1]
- Disruption of RNA Function: In parallel, 5-FU is also anabolized to 5-fluorouridine triphosphate (FUTP).[1] Due to its structural similarity to uridine triphosphate (UTP), FUTP is mistakenly recognized and incorporated into newly synthesized RNA strands by nuclear transcriptional enzymes. This fraudulent incorporation of a fluorinated base disrupts the structure, processing, and stability of multiple forms of RNA, including messenger RNA (mRNA) and ribosomal RNA (rRNA). The consequence is a cascade of downstream effects, including impaired protein synthesis and cellular dysfunction, which contributes significantly to the drug's overall cytotoxicity and induction of apoptosis.[1]
Pharmacokinetics: A Profile of Systemic Exposure and Disposition
The pharmacokinetic profile of capecitabine and its metabolites defines the drug's absorption, distribution, metabolism, and elimination (ADME), which collectively determine its therapeutic efficacy and toxicity profile. The journey from oral ingestion to elimination is a well-characterized process that underscores both the drug's rational design and the clinical considerations necessary for its safe use.
Absorption
Capecitabine is formulated for oral administration and is readily absorbed from the gastrointestinal tract.[4]
- Time to Peak Concentration (Tmax): Following a standard oral dose, the parent drug, capecitabine, reaches its maximum plasma concentration (Cmax) in approximately 1.5 hours. The key active metabolite, 5-FU, reaches its peak concentration slightly later, at approximately 2 hours post-administration.[4]
- Effect of Food: The presence of food has a significant impact on the absorption of capecitabine. Administration with a medium-fat, medium-carbohydrate meal decreases both the rate and extent of absorption. For the parent drug, mean Cmax is reduced by approximately 60% and the total exposure (Area Under the Curve, AUC0−∞) is reduced by 35%. For the active metabolite 5-FU, the Cmax and AUC0−∞ are also reduced by 43% and 21%, respectively. Food also delays the Tmax for both compounds by 1.5 hours.[4] Despite this reduction in bioavailability, clinical guidelines universally recommend that capecitabine be taken with food (specifically, within 30 minutes after a meal).[3] This seemingly paradoxical recommendation is a pragmatic clinical strategy that prioritizes tolerability. The blunting of peak plasma concentrations is believed to mitigate acute gastrointestinal side effects like nausea, while consistent administration with meals helps standardize the absorption profile, leading to more predictable drug exposure and a better overall safety profile in a real-world setting.
Distribution
Once absorbed, capecitabine and its metabolites are distributed throughout the body.
- Plasma Protein Binding: The binding of capecitabine and its metabolites to plasma proteins is low, at less than 60%, and is not dependent on drug concentration. Capecitabine itself binds primarily to human albumin, at a level of approximately 35%.[4] This low level of protein binding means a high fraction of the drug is free and available for distribution into tissues and subsequent metabolism.
- Tumor Penetration: A cornerstone of capecitabine's design is its ability to preferentially deliver its cytotoxic payload to malignant tissues. Clinical studies have validated this concept. In patients with colorectal cancer, the median concentration of the active metabolite 5-FU was found to be 2.9 times higher in tumor tissue compared to adjacent healthy tissue, with a range of 0.9 to 8.0.[7] This confirms the targeted drug delivery intended by the tumor-selective activation mechanism.
Metabolism
Capecitabine is extensively metabolized, first through the three-step activation cascade to 5-FU as detailed previously, and then through the catabolism of 5-FU itself.
- Catabolism of 5-FU: The activity of 5-FU is terminated by the enzyme dihydropyrimidine dehydrogenase (DPD), which is widely expressed in the body. DPD catalyzes the reduction of 5-FU to the far less toxic metabolite, 5-fluoro-5,6-dihydro-fluorouracil (FUH2). This is the first and rate-limiting step in the catabolic pathway. Subsequently, the pyrimidine ring of FUH2 is cleaved by dihydropyrimidinase to yield 5-fluoro-ureido-propionic acid (FUPA). Finally, FUPA is cleaved by β-ureido-propionase to form α-fluoro-β-alanine (FBAL), which is the major, inactive metabolite ultimately excreted in the urine.[4]
Elimination
The drug and its metabolites are cleared from the body relatively quickly.
- Elimination Half-Life (t1/2): Both the parent drug capecitabine and its active metabolite 5-FU have a short elimination half-life of approximately 0.75 hours.[4]
- Excretion: The primary route of elimination is renal. Following administration of a radiolabeled dose of capecitabine, 95.5% of the dose was recovered in the urine. Fecal excretion was minimal, accounting for only 2.6% of the dose. The vast majority of the excreted drug is in the form of metabolites, with the inactive FBAL being the most abundant, representing 57% of the administered dose. Only a very small fraction (3%) of the parent capecitabine is excreted unchanged.[4]
Pharmacokinetics in Special Populations
The pharmacokinetic behavior of capecitabine can be significantly altered in certain patient populations, necessitating careful monitoring and dose adjustments.
- Renal Impairment: This is the most critical patient-specific factor influencing capecitabine pharmacokinetics.
- Moderate Impairment (Creatinine Clearance [CLcr] 30-50 mL/min): Patients with moderate renal impairment exhibit significantly increased systemic exposure to the prodrug 5'-DFUR (AUC increased by 42%) and the inactive metabolite FBAL (AUC increased by 85%). This accumulation necessitates a 25% reduction in the starting dose of capecitabine to mitigate the risk of toxicity.[7]
- Severe Impairment (CLcr < 30 mL/min): In patients with severe renal impairment, exposure to metabolites is dramatically increased (e.g., FBAL AUC increased by 258%). A safe dosage has not been established for this population, and the use of capecitabine is generally contraindicated or not recommended.[7]
- Hepatic Impairment: In patients with mild to moderate hepatic impairment due to liver metastases, the total exposure (AUC) to the parent drug capecitabine was increased by 60%. However, the exposure to the active metabolite 5-FU was not significantly affected. Therefore, no starting dose adjustment is typically required for patients with mild-to-moderate hepatic impairment. The effects of severe hepatic impairment on capecitabine pharmacokinetics are unknown.[4]
- Other Populations: No clinically meaningful pharmacokinetic differences have been observed based on sex or between White and Black patient populations. While some differences in exposure were noted between Japanese and White patients, the clinical significance of these findings remains unknown.[7] In geriatric patients, a modest increase in the exposure to the inactive metabolite FBAL has been observed, which is likely attributable to the natural age-related decline in renal function.[7]
The complete pharmacokinetic pathway highlights two critical points of control for drug exposure and potential toxicity. The first is the catabolism of 5-FU by DPD, where genetic deficiencies can lead to a "traffic jam" of the active drug, dramatically increasing toxicity risk. The second is renal function, which is responsible for clearing the drug's metabolites. Impairment in this function can lead to their accumulation. This underscores why baseline assessment of renal function (for CLcr calculation) and, where feasible, screening for DPD deficiency are paramount safety measures before initiating capecitabine therapy.
Parameter | Capecitabine | Fluorouracil (5-FU) | Source(s) |
---|
Tmax (Fasting) | ~1.5 hours | ~2.0 hours | 4 |
Effect of Food on Cmax | ↓ 60% | ↓ 43% | 4 |
Effect of Food on AUC | ↓ 35% | ↓ 21% | 4 |
Plasma Protein Binding | ~35% (to albumin) | <60% | 4 |
Elimination Half-Life | ~0.75 hours | ~0.75 hours | 4 |
Primary Excretion Route | Renal (95.5% of dose recovered in urine) | - | 4 |
Table 2: Summary of Key Pharmacokinetic Parameters for Capecitabine and its Active Metabolite 5-FU | | | |
Population | Creatinine Clearance (CLcr) | Recommended Dose Adjustment | Source(s) |
---|
Normal Renal Function | > 50 mL/min | No adjustment needed | 7 |
Moderate Renal Impairment | 30 to 50 mL/min | Reduce starting dose by 25% | 7 |
Severe Renal Impairment | < 30 mL/min | Use is not recommended / contraindicated | 7 |
Table 3: Recommended Dose Adjustments for Renal Impairment | | | |
Clinical Efficacy and Approved Therapeutic Indications
Capecitabine has demonstrated robust clinical efficacy across a range of solid tumors, leading to its approval by major regulatory bodies, including the FDA and EMA, and its establishment as a standard of care in multiple therapeutic settings.[1] Its development trajectory is a classic example of a successful oncology agent, initially proving its value in advanced, refractory disease and subsequently moving into earlier lines of therapy and adjuvant settings based on strong clinical trial evidence. This progression underscores its favorable balance of efficacy and tolerability.
Colorectal Cancer (CRC)
Capecitabine plays a pivotal role in the management of colorectal cancer, from early-stage to metastatic disease.
- Adjuvant Treatment of Stage III (Dukes' C) Colon Cancer: Following surgical resection, capecitabine is indicated for adjuvant therapy to reduce the risk of recurrence. It can be used either as a single agent or as a component of a combination regimen with oxaliplatin (a regimen known as CAPOX or XELOX).[1] Post-marketing phase 4 studies, such as NCT00502671 and NCT02581423, have further supported its role in this curative-intent setting.[29]
- Metastatic Colorectal Cancer (mCRC): For patients with unresectable or metastatic disease, capecitabine is indicated as a first-line treatment, both as monotherapy (particularly when fluoropyrimidine therapy alone is preferred) and as the fluoropyrimidine backbone in various combination regimens.[1] The OBELIX trial (NCT00577031), for instance, investigated its use in combination with oxaliplatin and bevacizumab for mCRC.[29]
- Perioperative Treatment of Rectal Cancer: In the management of locally advanced rectal cancer, capecitabine is used as a component of neoadjuvant (preoperative) chemoradiotherapy, where it acts as a radiosensitizer to improve tumor response prior to surgery.[1]
Breast Cancer
Capecitabine is a key therapeutic option for patients with advanced or metastatic breast cancer (MBC).
- Monotherapy: It is indicated as a single agent for the treatment of patients with MBC whose disease is resistant to or has progressed following prior chemotherapy regimens containing both a taxane (e.g., paclitaxel) and an anthracycline (e.g., doxorubicin). It is also an option for patients for whom further anthracycline therapy is not indicated.[1]
- Combination Therapy: Capecitabine is approved for use in combination with docetaxel for the treatment of patients with MBC after failure of a prior anthracycline-containing chemotherapy regimen.[1]
- Clinical Trial Evidence: The XEPAD study (NCT01725386) was a large observational trial designed to evaluate the routine clinical use, safety, and efficacy of capecitabine in the MBC setting.[30] More recently, the phase III MECCA trial demonstrated a significant improvement in both progression-free survival (PFS) and overall survival (OS) when metronomic (low-dose, continuous) capecitabine was added to an aromatase inhibitor as a first-line treatment for patients with hormone receptor-positive (HR+), HER2-negative MBC.[1]
Gastric, Esophageal, or Gastroesophageal Junction (GEJ) Cancer
Capecitabine has replaced intravenous 5-FU in many combination regimens for upper gastrointestinal cancers due to its convenience and comparable efficacy.
- It is indicated as a component of combination chemotherapy for the first-line treatment of adults with unresectable or metastatic gastric, esophageal, or GEJ cancer.[1]
- For patients with HER2-overexpressing metastatic gastric or GEJ adenocarcinoma who have not received prior therapy for metastatic disease, capecitabine is specifically indicated for use in a triplet regimen with cisplatin and trastuzumab.[1]
Pancreatic Cancer
In the challenging landscape of pancreatic cancer treatment, capecitabine has carved out a role in the adjuvant setting.
- It is indicated for the adjuvant treatment of adult patients with pancreatic adenocarcinoma as a component of a combination chemotherapy regimen with gemcitabine.[1]
The widespread adoption and regulatory success of capecitabine marked a significant shift in oncology practice, demonstrating that a well-designed oral agent could effectively replace or offer a non-inferior alternative to a standard-of-care intravenous drug. This paradigm shift has had a profound impact on the delivery of cancer care, affording patients greater convenience, less time committed to hospital visits, and increased autonomy over their treatment, all of which contribute to an improved quality of life during therapy.
Dosing Regimens, Administration, and Formulation
The clinical application of capecitabine requires a precise understanding of its complex dosing, administration guidelines, and available formulations. Unlike many oral medications with a simple fixed dose, capecitabine dosing is individualized based on body surface area (BSA) and indication, often requiring patients to take a combination of tablet strengths on an intermittent cycle. This complexity underscores the importance of careful calculation by clinicians and education for patients to ensure both safety and efficacy.
Recommended Dosing by Indication
The recommended dosage of capecitabine varies by indication, whether it is used as monotherapy or in combination, and the specific treatment cycle.
Indication | Regimen | Recommended Dosage and Schedule | Source(s) |
---|
Adjuvant Colon Cancer | Monotherapy | 1,250 mg/m² orally twice daily on Days 1-14 of a 21-day cycle, for 8 cycles. | 7 |
| Combination (CAPOX) | 1,000 mg/m² orally twice daily on Days 1-14 of a 21-day cycle, for 8 cycles. | 7 |
Metastatic CRC | Monotherapy | 1,250 mg/m² orally twice daily on Days 1-14 of a 21-day cycle. | 7 |
Metastatic Breast Cancer | Monotherapy or Combination | 1,000 to 1,250 mg/m² orally twice daily on Days 1-14 of a 21-day cycle. | 7 |
Gastric/Esophageal/GEJ Cancer | Combination | 850 to 1,000 mg/m² orally twice daily on Days 1-14 of a 21-day cycle, OR 625 mg/m² twice daily continuously. | 7 |
Adjuvant Pancreatic Cancer | Combination (with Gemcitabine) | 830 mg/m² orally twice daily on Days 1-21 of a 28-day cycle, for 6 cycles. | 7 |
Table 4: Approved Dosage Regimens by Indication | | | |
Dosage Modifications for Adverse Reactions
Proactive management of toxicities is crucial for maintaining patients on therapy. The FDA label provides a clear dose modification scheme based on the National Cancer Institute of Canada (NCIC) grading of adverse events.
- Grade 2 Toxicity: For the first occurrence, treatment should be interrupted until the event resolves to Grade 0-1, then resumed at 100% of the current dose. For a second occurrence, the dose should be reduced to 75%. For a third occurrence, the dose is reduced to 50%. A fourth occurrence necessitates permanent discontinuation.
- Grade 3 Toxicity: For the first occurrence, treatment should be interrupted until resolution and then resumed at a 75% dose. For a second occurrence, the dose is reduced to 50%. Treatment should be permanently discontinued upon a third occurrence.
- Grade 4 Toxicity: Treatment should be permanently discontinued. Alternatively, if it is in the physician's judgment to be in the patient's best interest, treatment can be interrupted until resolution and then resumed at a 50% dose.[7]
Administration Guidelines
Proper administration is essential for safety and consistency of drug exposure.
- With Food: Tablets should be swallowed whole with water within 30 minutes after the end of a meal.[7]
- Timing: Doses should be taken approximately 12 hours apart to maintain steady drug levels.[1]
- Tablet Integrity: Patients must be instructed to swallow tablets whole. They should not be cut, crushed, or chewed, as this can alter absorption and poses an exposure risk to the patient and caregivers.[7] If a tablet must be cut or crushed, this should only be performed by a trained professional (e.g., a pharmacist) using appropriate safety equipment and procedures.[7]
- Missed or Vomited Doses: If a dose is missed or vomited, the patient should not take a replacement dose. They should simply skip that dose and continue with the next scheduled dose at the regular time.[7]
Formulations and Visual Identification
Capecitabine is available in two strengths to facilitate BSA-based dosing. Accurate identification is critical to prevent medication errors.
- Available Strengths: Film-coated tablets are available in 150 mg and 500 mg strengths.[4]
- Visual Appearance:
- Brand (Xeloda®): The brand-name tablets have a distinctive appearance. The 150 mg tablet is light-peach, biconvex, and oblong, imprinted with "XELODA" on one side and "150" on the other. The 500 mg tablet is a darker peach color but otherwise similar in shape, imprinted with "XELODA" and "500".[4]
- Generic Formulations: Generic versions are produced by many manufacturers and, while generally peach-colored and oblong/oval, they feature different imprints. This variation can be a source of confusion for patients. Examples of imprints on generic 500 mg tablets include "RDY 500", "A016 500", "C500", and "H 3".[33]
The inherent complexity of capecitabine's dosing—requiring BSA calculations, combinations of different tablet strengths, and intermittent cycling—places a significant cognitive load on both healthcare providers and patients, creating multiple opportunities for medication errors. This practical challenge has been a primary motivator for the extensive clinical research into simplified, fixed-dose, or alternative schedules, which aim to improve safety and ease of use without compromising efficacy.[9]
Formulation | Strength | Color | Shape | Imprint | Source(s) |
---|
Xeloda® (Brand) | 150 mg | Light Peach | Biconvex, Oblong | XELODA / 150 | 34 |
| 500 mg | Peach | Biconvex, Oblong | XELODA / 500 | 34 |
Generic Example 1 | 150 mg | Peach | Capsule/Oblong | RDY 150 | 36 |
| 500 mg | Peach | Capsule/Oblong | RDY 500 | 36 |
Generic Example 2 | 150 mg | Peach | Oval | A015 150 | 36 |
| 500 mg | Peach | Oval | A016 500 | 36 |
Generic Example 3 | 500 mg | Peach | Oval | H 3 | 36 |
Table 5: Visual Identification Guide for Common Capecitabine Formulations | | | | | |
Safety, Tolerability, and Risk Management
The clinical use of capecitabine is guided by its well-characterized but significant safety profile. While generally considered manageable, its toxicities can be severe and require vigilant monitoring, patient education, and proactive dose modifications. The safety profile is a direct reflection of its pharmacodynamic mechanism, mimicking the effects of continuous 5-FU infusion.
Boxed Warning: Warfarin Interaction
The U.S. FDA has issued a boxed warning for capecitabine, its most serious level of warning, regarding a clinically significant drug interaction with coumarin-derivative anticoagulants such as warfarin.[4]
- Risk: Concomitant use can lead to altered coagulation parameters (clinically significant increases in International Normalized Ratio or Prothrombin Time) and subsequent bleeding events, which have been severe and in some cases, fatal.
- Monitoring: Patients receiving this combination must have their anticoagulant response monitored frequently (more often than usual) to allow for appropriate and timely adjustment of the anticoagulant dose.
- Onset: These events can occur within days to several months of initiating capecitabine and have even been reported within one month of its discontinuation. The risk is present in patients both with and without liver metastases.[4]
Contraindications
The use of capecitabine is strictly contraindicated in the following populations:
- Patients with a history of a severe hypersensitivity reaction to capecitabine or its active metabolite, 5-fluorouracil.[7]
- Patients with a known complete deficiency of the enzyme dihydropyrimidine dehydrogenase (DPD).[7]
Warnings and Precautions
Several major risks associated with capecitabine therapy require careful management.
- Dihydropyrimidine Dehydrogenase (DPD) Deficiency: This is a critical pharmacogenomic consideration. DPD is the primary enzyme responsible for the catabolism and inactivation of 5-FU. Patients with partial or complete DPD deficiency, caused by genetic variants in the DPYD gene, are unable to clear the active drug effectively. This leads to massively increased systemic exposure to 5-FU and a dramatically elevated risk of acute, severe, and potentially fatal toxicities, including severe mucositis, diarrhea, neutropenia, and neurotoxicity.[7] This risk highlights a major advancement in oncologic safety: the shift toward proactive, predictive risk mitigation. Pre-treatment genotyping for
DPYD variants is now recommended or standard practice in many regions to identify at-risk patients, allowing for dose reduction or selection of an alternative therapy, thereby moving away from a purely reactive model of toxicity management.[7]
- Cardiotoxicity: Capecitabine can induce cardiotoxicity, with manifestations ranging from ECG changes to angina, dysrhythmias, myocardial infarction/ischemia, cardiac failure, and sudden death. The risk is elevated in patients with a pre-existing history of coronary artery disease. If cardiotoxicity occurs, the drug must be withheld, and the safety of resumption has not been established.[7]
- Gastrointestinal Toxicity: Severe diarrhea is a very common and dose-limiting toxicity. It must be managed promptly with antidiarrheal medication and dose interruption/reduction. Uncontrolled diarrhea can lead to severe dehydration and subsequent acute renal failure.[7]
- Dermatologic Toxicity (Palmar-Plantar Erythrodysesthesia): Also known as Hand-Foot Syndrome (HFS), PPES is one of the most characteristic and frequent toxicities of capecitabine. It presents with a range of symptoms on the palms of the hands and soles of the feet, including redness, swelling, blistering, pain, and numbness. In severe cases, it can be debilitating and is a primary reason for dose modification or discontinuation. Persistent, severe PPES can even lead to the loss of fingerprints.[7] The prevalence of this specific toxicity is a direct clinical manifestation of the drug's intended mechanism, which mimics continuous 5-FU exposure.
- Myelosuppression: Treatment can lead to bone marrow suppression, resulting in neutropenia, thrombocytopenia, and anemia. Complete blood counts must be monitored at baseline and before each treatment cycle. Necrotizing enterocolitis (typhlitis), a serious complication, has also been reported.[7]
- Hyperbilirubinemia: Significant elevations in bilirubin can occur, particularly in patients with hepatic metastases. Liver function tests should be monitored, and dose modifications are required for Grade 3-4 hyperbilirubinemia.[7]
- Embryo-Fetal Toxicity: Capecitabine is classified as a teratogenic agent and can cause fetal harm when administered during pregnancy. Females of reproductive potential must be advised to use effective contraception during treatment and for 6 months after the final dose. Males with female partners of reproductive potential should use effective contraception during treatment and for 3 months after the final dose.[7]
- Ocular Toxicity: Post-marketing surveillance has identified ocular adverse events, including lacrimal duct stenosis (narrowing of the tear duct) and corneal disorders such as keratitis.[7]
Common and Serious Adverse Reactions
The incidence of adverse reactions varies depending on whether capecitabine is used as a single agent or in combination with other cytotoxic drugs.
Adverse Reaction | Adjuvant Colon Cancer (Monotherapy) | Metastatic Breast Cancer (Monotherapy) | Metastatic Breast Cancer (with Docetaxel) |
---|
| All Grades | Grade 3/4 | All Grades |
Palmar-Plantar Erythrodysesthesia | 60% | 17% | 57% |
Diarrhea | 47% | 12% | 57% |
Nausea | 34% | 2% | 53% |
Stomatitis | 22% | 2% | 25% |
Vomiting | 21% | 2% | 37% |
Fatigue/Asthenia | 21% | 3% | 41% |
Neutropenia | N/A | <1% | N/A |
Anemia | 72% | 1% | 72% |
Hyperbilirubinemia | 23% | 11% | 22% |
Data derived from FDA prescribing information. N/A indicates data not reported at ≥20% threshold for all grades in that specific trial. | | | |
Table 6: Incidence of Common Adverse Reactions (≥20% All Grades) in Pivotal Trials 7 | | | |
Clinically Significant Drug Interactions
The safe use of capecitabine requires careful consideration of potential drug-drug interactions. While its metabolic pathway does not heavily rely on the cytochrome P450 system, its most critical interactions are pharmacodynamic or involve its specific catabolic enzyme, DPD.
Interaction with Vitamin K Antagonists (Warfarin)
This is the most significant and well-documented interaction, carrying a boxed warning from the FDA.[4] Co-administration of capecitabine with warfarin or other coumarin-derivative anticoagulants can potentiate the anticoagulant effect, leading to clinically significant increases in INR and a high risk of severe, potentially fatal bleeding. The exact mechanism is not fully understood but is considered pharmacodynamic. Frequent and vigilant monitoring of INR or prothrombin time is mandatory for any patient on this combination, with prompt dose adjustments of the anticoagulant as needed.[1]
Interaction with DPD-Inhibiting Drugs
The enzyme DPD is essential for the inactivation of 5-FU. Co-administration of capecitabine with potent DPD inhibitors can lead to a massive accumulation of 5-FU and life-threatening toxicity. The antiviral agent sorivudine and its chemically related analogues (e.g., brivudine) are strong DPD inhibitors, and their concomitant use with capecitabine is contraindicated.[1]
Interaction with Phenytoin
Capecitabine can increase the plasma concentrations of phenytoin. For patients receiving these drugs concomitantly, regular monitoring of phenytoin concentrations is recommended to avoid toxicity.[1] The mechanism is thought to involve inhibition of the CYP2C9 enzyme, which metabolizes phenytoin.
Interaction with Leucovorin / Folic Acid
Leucovorin (folinic acid) and high doses of folic acid supplements can enhance the toxicity of 5-FU. Leucovorin's active metabolite stabilizes the binding of FdUMP to the target enzyme, thymidylate synthase, thereby increasing the potency and duration of its inhibitory effect. While leucovorin is intentionally combined with 5-FU in some regimens (e.g., FOLFOX) to boost efficacy, patients taking capecitabine should be advised to report the use of any folic acid-containing supplements to their physician, as this can unintentionally increase the risk of toxicity.[1]
Interaction with Nephrotoxic Agents
Because capecitabine's metabolites are cleared by the kidneys, co-administration with other drugs that can impair renal function (nephrotoxic agents) may increase the risk of capecitabine-related toxicity. Caution is advised when combining capecitabine with drugs known to be nephrotoxic.[1]
Interaction with Vaccines
As an immunosuppressive agent, capecitabine can reduce the host's immune response to vaccines, potentially diminishing their efficacy. Furthermore, the administration of live or live-attenuated vaccines to patients receiving capecitabine is not recommended due to the increased risk of inducing a disseminated infection.[1]
Drug-Food Interaction
As detailed in the Pharmacokinetics section, food decreases the rate and extent of capecitabine absorption.[4] However, to improve gastrointestinal tolerability, administration with food (within 30 minutes of a meal) is the standard clinical recommendation.[1]
The Evolving Clinical Landscape: A Review of Clinical Trials and Off-Label Applications
Capecitabine, despite being a mature drug approved over two decades ago, remains the subject of extensive and dynamic clinical investigation.[10] This ongoing research reflects the lifecycle of a successful oncology agent, where investigation expands beyond initial approvals into three key domains: 1) indication expansion through off-label use, 2) optimization of existing therapy through novel dosing schedules, and 3) synergistic combination with the next generation of cancer treatments.
Off-Label Use and Guideline-Endorsed Indications
While capecitabine has specific FDA-approved indications, its utility extends to a wide array of other solid malignancies. This off-label use is frequently supported by robust clinical data and endorsement in major clinical practice guidelines, such as those from the National Comprehensive Cancer Network (NCCN).[27] Documented and guideline-supported off-label applications include:
- Anal Squamous Cell Carcinoma [27]
- Neuroendocrine Tumors (including of the pancreas) [28]
- Ovarian, Fallopian Tube, and Primary Peritoneal Cancer [8]
- Small Bowel Adenocarcinoma [28]
- Thymomas and Thymic Carcinomas [28]
- Nasopharyngeal Carcinoma [40]
- Gynecological Cancers [41]
- Penile Cancer and Squamous Cell Skin Cancer [28]
Exploration of Alternative Dosing Schedules
A major focus of modern clinical research on capecitabine is the optimization of its dosing schedule. This effort is driven by the central challenge of chemotherapy: balancing efficacy against treatment-related toxicity and its impact on patient quality of life. The standard 14-days-on, 7-days-off regimen is effective but is associated with a high incidence of dose-limiting toxicities, particularly palmar-plantar erythrodysesthesia (HFS) and diarrhea, which often necessitate dose reductions.[9] This has led to the hypothesis that the "maximum tolerated dose" (MTD) paradigm may not be optimal, and that alternative schedules might achieve a better therapeutic index. Key alternative regimens under investigation include:
- 7-Days-On, 7-Days-Off Schedule: Several trials, including the randomized phase II study NCT02595320, are comparing a fixed-dose, 7-on/7-off schedule against the standard BSA-based 14/7 schedule. The primary hypothesis is that this dose-dense but shorter-duration schedule will result in similar efficacy with significantly less toxicity.[9]
- Metronomic Dosing: This approach involves administering a lower dose of the drug on a more continuous or frequent schedule. The phase III MECCA trial provided strong evidence for this strategy, showing that the addition of metronomic capecitabine to an aromatase inhibitor significantly improved survival outcomes in HR+/HER2- metastatic breast cancer.[1]
- Continuous Dosing: The trial NCT00418028 explored a continuous low-dose capecitabine schedule (800 mg/m²/day) without a rest period, with the specific goal of reducing the incidence of Grade ≥2 HFS from an expected 50% on the standard arm to 20%.[25]
Novel Combination Therapies
Capecitabine's established efficacy, oral convenience, and manageable safety profile make it an ideal backbone for combination with novel therapeutic agents.
- Immunotherapy Combinations: There is emerging preclinical evidence that fluoropyrimidines may have immunomodulatory effects, such as increasing the expression of PD-L1 on tumor cells. This provides a strong rationale for combining capecitabine with immune checkpoint inhibitors. The phase I trial NCT05064085, for example, evaluated the safety and efficacy of capecitabine sequenced with the PD-1 inhibitor cemiplimab in metastatic breast cancer, reporting an acceptable safety profile and a promising clinical benefit rate.[42]
- Targeted Therapy Combinations: Capecitabine is being actively studied in combination with a wide range of targeted agents. The National Cancer Institute (NCI) trial portfolio lists numerous active studies combining capecitabine with drugs targeting various pathways, such as PARP inhibitors (e.g., ZEN003694), HER2-targeted agents (e.g., zanidatamab), and antibody-drug conjugates (e.g., sacituzumab govitecan).[43]
The breadth of ongoing research, from exploring new indications in gynecological cancers to its use as a standard comparator arm in trials for new colorectal and breast cancer drugs, confirms that capecitabine is far from a static or obsolete therapy. Instead, it is a dynamic tool whose full potential is still being unlocked through intelligent dose optimization and synergistic combination strategies.
Expert Analysis and Strategic Outlook
Capecitabine stands as a paradigm of successful rational drug design in oncology. Its creation was a direct and effective effort to translate the proven efficacy of continuous intravenous infusion 5-fluorouracil into a convenient, orally administered prodrug with a potentially improved therapeutic index. Over two decades since its initial approval, it has firmly established itself as a "workhorse" chemotherapeutic, forming the backbone of standard-of-care treatment for several of the world's most prevalent cancers and frequently serving as the benchmark comparator arm against which new therapies are evaluated.[10]
Key Strengths and Limitations
The enduring clinical relevance of capecitabine is built upon a unique combination of strengths, balanced against a set of well-defined limitations that guide its clinical use.
- Strengths: The primary advantage is its oral route of administration, which profoundly enhances patient convenience and quality of life. This is coupled with a mechanism of tumor-selective activation, which aims to concentrate its cytotoxic effect at the site of malignancy. Its robust efficacy is proven across multiple indications and lines of therapy, and its toxicity profile, while significant, is well-characterized, predictable, and generally manageable with proactive monitoring and established dose-modification protocols.
- Limitations: The drug is associated with significant and often dose-limiting toxicities, most notably palmar-plantar erythrodysesthesia (HFS) and diarrhea. Its complex, body-surface-area-based dosing creates potential for calculation and administration errors. A critical limitation is the risk of severe toxicity stemming from pharmacogenomic variability in the DPD enzyme, making DPD deficiency a crucial safety consideration. Finally, its dangerous interaction with warfarin necessitates extreme caution and vigilant monitoring in patients requiring anticoagulation.
Future Perspectives and Unanswered Questions
The future of capecitabine in an era of precision medicine and immunotherapy will be defined not by questions of its intrinsic efficacy, but by how to use it more intelligently, safely, and synergistically.
- The Future of Dosing: A pivotal question is whether alternative dosing schedules, such as the 7-days-on, 7-days-off regimen, will replace the current standard of care. The results of ongoing randomized trials are highly anticipated, as a positive outcome could fundamentally change prescribing practices, offering a regimen with a potentially superior balance of efficacy and tolerability.
- Biomarker-Driven Therapy: The principle of tumor-selective activation via thymidine phosphorylase (dThdPase) has long suggested a potential role for biomarkers. A key unanswered question is whether tumor dThdPase expression levels or other molecular markers will ever be prospectively validated to select patients most likely to respond, thereby moving beyond empirical use to a more personalized approach.
- Role in the Immunotherapy Era: Early data suggesting that capecitabine may have immunomodulatory properties is intriguing. Larger, randomized trials are needed to determine if it can be effectively repurposed as a synergistic partner for immune checkpoint inhibitors, potentially sensitizing "cold" tumors to immunotherapy and expanding the population of patients who can benefit from these transformative agents.
Concluding Remarks
In conclusion, capecitabine remains an indispensable tool in clinical oncology. Its journey from a rationally designed prodrug to a global standard of care is a testament to its success. While the landscape of cancer therapy is rapidly evolving with novel targeted agents and immunotherapies, capecitabine is not being replaced but rather integrated and re-examined. The focus of current and future research—optimizing its tolerability through novel scheduling and harnessing its potential in new combinations—ensures that this mature drug will retain its clinical value and continue to play a vital role in the treatment of cancer for the foreseeable future.
Works cited
- Capecitabine: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed July 16, 2025, https://go.drugbank.com/drugs/DB01101
- Capecitabine - Wikipedia, accessed July 16, 2025, https://en.wikipedia.org/wiki/Capecitabine
- Buy Generic Xeloda (Capecitabine) 500mg for Breast Cancer, accessed July 16, 2025, https://www.breastcancerhelpinc.org/xeloda-capecitabine
- Xeloda (capecitabine) tablets Label - accessdata.fda.gov, accessed July 16, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2005/020896s016lbl.pdf
- capecitabine | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed July 16, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=6799
- Capecitabine | C15H22FN3O6 | CID 60953 - PubChem, accessed July 16, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Capecitabine
- XELODA® (capecitabine) tablets, for oral use - accessdata.fda.gov, accessed July 16, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020896s044s045s046s047s048s049s050s051lbl.pdf
- A phase II trial of capecitabine (Xeloda®) in recurrent ovarian cancer - PMC, accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2394434/
- Randomized Trial of Fixed-Dose Capecitabine Compared With Standard-Dose Capecitabine in Metastatic Breast Cancer: X-7/7 Trial | JCO Oncology Advances - ASCO Publications, accessed July 16, 2025, https://ascopubs.org/doi/10.1200/OA-24-00068
- Capecitabine monotherapy as first-line treatment in advanced HER2-normal breast cancer – a nationwide, retrospective study - PubMed Central, accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11332473/
- Study Details | Capecitabine in Metastatic Breast and GI Cancers | ClinicalTrials.gov, accessed July 16, 2025, https://clinicaltrials.gov/ct2/show/NCT02595320
- Capecitabine | CAS#154361-50-9 | antimetabolite - MedKoo Biosciences, accessed July 16, 2025, https://www.medkoo.com/products/4614
- Capecitabine (T3D3507) - T3DB, accessed July 16, 2025, https://t3db.ca/toxins/T3D3507
- Capecitabine (Ro 09-1978, CAS Number: 154361-50-9) | Cayman Chemical, accessed July 16, 2025, https://www.caymanchem.com/product/10487/capecitabine
- Capecitabine | CAS 154361-50-9 | SCBT - Santa Cruz Biotechnology, accessed July 16, 2025, https://www.scbt.com/p/capecitabine-154361-50-9
- [Capecitabine (200 mg)] - CAS [154361-50-9] - USP Store, accessed July 16, 2025, https://store.usp.org/product/1090706
- Capecitabine = 98 HPLC 154361-50-9 - Sigma-Aldrich, accessed July 16, 2025, https://www.sigmaaldrich.com/US/en/product/sigma/sml0653
- Capecitabine | 99.93%(HPLC) | In Stock | DNA/RNA Synthesis inhibitor - Selleck Chemicals, accessed July 16, 2025, https://www.selleckchem.com/products/Capecitabine(Xeloda).html
- Capecitabine (Standard) | DNA/RNA Synthesis Inhibitor | MedChemExpress, accessed July 16, 2025, https://www.medchemexpress.com/capecitabine-standard.html
- Capecitabine | C15H22FN3O6 - ChemSpider, accessed July 16, 2025, https://www.chemspider.com/Chemical-Structure.54916.html
- Definition of capecitabine - NCI Drug Dictionary, accessed July 16, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/capecitabine
- Compound: CAPECITABINE (CHEMBL1773) - ChEMBL - EMBL-EBI, accessed July 16, 2025, https://www.ebi.ac.uk/chembl/explore/compound/CHEMBL1773
- www.researchgate.net, accessed July 16, 2025, https://www.researchgate.net/figure/Capecitabine-chemical-structure-Molecular-weight-359350083-G-mo-Molecular_fig3_393440344#:~:text=NonCommercial%204.0%20International-,Capecitabine%20chemical%20structure%20%EF%82%B7%20Molecular%20weight%3A%20359.350083%20(G%2F,mo)%20%EF%82%B7%20Molecular%20Formula%3A%20C13H24EN20&text=Nanoparticle%20based%20drugs%20are%20an,minimising%20damage%20to%20healthy%20cells.
- Capecitabine chemical structure Molecular weight: 359.350083 (G/mo) - ResearchGate, accessed July 16, 2025, https://www.researchgate.net/figure/Capecitabine-chemical-structure-Molecular-weight-359350083-G-mo-Molecular_fig3_393440344
- Study Details | Standard Versus Continuous Capecitabine in Advanced Breast Cancer, accessed July 16, 2025, https://www.clinicaltrials.gov/study/NCT00418028
- Capecitabine (oral route) - Mayo Clinic, accessed July 16, 2025, https://www.mayoclinic.org/drugs-supplements/capecitabine-oral-route/description/drg-20062501
- Clinical Policy: Capecitabine (Xeloda) - PA Health & Wellness, accessed July 16, 2025, https://www.pahealthwellness.com/content/dam/centene/Pennsylvania/policies/pharmacy-policies/PA.CP.PHAR.60%20Capecitabine.pdf
- Clinical Policy: Capecitabine (Xeloda) - Superior Health Plan, accessed July 16, 2025, https://www.superiorhealthplan.com/content/dam/centene/Superior/policies/pharmacy-policies/CP.PHAR.60-06012021.pdf
- CRC Completed Phase 4 Trials for Capecitabine (DB01101) | DrugBank Online, accessed July 16, 2025, https://go.drugbank.com/indications/DBCOND0051083/clinical_trials/DB01101?phase=4&status=completed
- An Observational Study of Xeloda (Capecitabine) in Participants With Metastatic or Advanced Breast Cancer | ClinicalTrials.gov, accessed July 16, 2025, https://www.clinicaltrials.gov/study/NCT01725386
- Metronomic Capecitabine Plus Aromatase Inhibitor as Initial Therapy in Patients With Hormone Receptor–Positive, Human Epidermal Growth Factor Receptor 2–Negative Metastatic Breast Cancer: The Phase III MECCA Trial - ASCO Publications, accessed July 16, 2025, https://ascopubs.org/doi/10.1200/JCO.24.00938
- Medication Info Sheet: Capecitabine (Xeloda®) (NYULH), accessed July 16, 2025, https://nyulangoneib.staywellsolutionsonline.com/Library/Encyclopedia/22,91109
- Generic Xeloda Availability - Drugs.com, accessed July 16, 2025, https://www.drugs.com/availability/generic-xeloda.html
- Xeloda Pill Images - What does Xeloda look like? - Drugs.com, accessed July 16, 2025, https://www.drugs.com/image/xeloda-images.html
- Label: Xeloda—capecitabine tablet, film coated - DailyMed, accessed July 16, 2025, https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=a1de8bba-3b1d-4c9d-ab8a-32d2c05e67c8
- Capecitabine Pill Images - What does capecitabine look like? - Drugs.com, accessed July 16, 2025, https://www.drugs.com/image/capecitabine-images.html
- Capecitabine (Xeloda) | Breast Cancer Now, accessed July 16, 2025, https://breastcancernow.org/about-breast-cancer/treatment/chemotherapy/chemotherapy-drugs/capecitabine-xeloda
- Xeloda (capecitabine) dosing, indications, interactions, adverse effects, and more, accessed July 16, 2025, https://reference.medscape.com/drug/xeloda-capecitabine-342211
- Tolerability, dose intensity, and prescribing patterns of capecitabine by race., accessed July 16, 2025, https://ascopubs.org/doi/10.1200/JCO.2024.42.16_suppl.e23278
- NPC Completed Phase 3 Trials for Capecitabine (DB01101) | DrugBank Online, accessed July 16, 2025, https://go.drugbank.com/indications/DBCOND0060814/clinical_trials/DB01101?phase=3&status=completed
- Gynecological Cancers Recruiting Phase 1 Trials for Capecitabine (DB01101) - DrugBank, accessed July 16, 2025, https://go.drugbank.com/indications/DBCOND0044877/clinical_trials/DB01101?phase=1&status=recruiting
- Phase I trial of capecitabine with cemiplimab in patients with hormone receptor–positive metastatic breast cancer. | Journal of Clinical Oncology - ASCO Publications, accessed July 16, 2025, https://ascopubs.org/doi/10.1200/JCO.2025.43.16_suppl.e13082
- Clinical Trials Using Capecitabine - NCI, accessed July 16, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/capecitabine?pn=1