C9H11F2N3O4
95058-81-4
Advanced Ovarian Cancer, Bladder Transitional Cell Carcinoma Stage IV, Carcinoma of the Head and Neck, Cervical Cancer, Cutaneous T-Cell Lymphoma (CTCL), Hodgkins Disease (HD), Mesothelioma, Metastatic Breast Cancer, Pancreatic Adenocarcinoma Locally Advanced, Small Cell Lung Cancer (SCLC), Stage IIIA Non Small Cell Lung Cancer, Stage IIIB Non-Small Cell Lung Cancer, Stage IV Non-small Cell Lung Cancer (NSCLC), Stage 4 Pancreatic adenocarcinoma
Gemcitabine is a synthetic pyrimidine nucleoside analog that functions as a cornerstone antimetabolite chemotherapeutic agent in modern oncology.[1] First synthesized in the 1980s and receiving its initial U.S. Food and Drug Administration (FDA) approval in 1996, it has become an indispensable treatment for a range of solid tumors.[3] Marketed under the originator brand name Gemzar® and numerous generic formulations, Gemcitabine is classified as a small molecule prodrug that requires intracellular activation to exert its cytotoxic effects.[4]
The drug's mechanism of action is distinguished by a dual and synergistic process. Following active transport into the cell and sequential phosphorylation to its active diphosphate (dFdCDP) and triphosphate (dFdCTP) forms, Gemcitabine disrupts cellular replication through two pathways. The triphosphate metabolite, dFdCTP, is incorporated into DNA, leading to an irreparable error known as "masked chain termination." Concurrently, the diphosphate metabolite, dFdCDP, inhibits ribonucleotide reductase, the enzyme responsible for generating the deoxynucleotides necessary for DNA synthesis. This inhibition depletes the natural competitor of dFdCTP, creating a self-potentiating cycle that enhances the drug's cytotoxic potency.[4]
Gemcitabine holds regulatory approval from major agencies, including the FDA and the European Medicines Agency (EMA), for numerous indications. It is a standard of care, typically in combination regimens, for pancreatic adenocarcinoma, non-small cell lung cancer (NSCLC), metastatic breast cancer, advanced ovarian cancer, and bladder cancer.[7] Its primary dose-limiting toxicity is myelosuppression, manifesting as neutropenia, anemia, and thrombocytopenia, which necessitates careful monitoring and dose adjustments.[4]
The therapeutic role of Gemcitabine continues to evolve. Initially established as a key monotherapy and a synergistic partner for traditional cytotoxic agents like platinum compounds and taxanes, its utility has been redefined in the era of immunotherapy. Recent landmark approvals for its use in combination with immune checkpoint inhibitors for biliary tract and urothelial cancers have highlighted its immunomodulatory properties, positioning it as a critical agent that can prime the tumor microenvironment for an effective anti-tumor immune response.[9] Ongoing research into novel formulations, resistance mechanisms, and biomarker-driven applications ensures that Gemcitabine will remain a vital component of cancer therapy for the foreseeable future.
The precise identification and characterization of a pharmaceutical agent's physicochemical properties are fundamental to understanding its formulation, pharmacokinetics, and clinical behavior. Gemcitabine is well-defined by a comprehensive set of chemical identifiers and experimental data.
Gemcitabine is known by several names and is cataloged in major chemical and pharmacological databases.
The clinical formulation and biological activity of Gemcitabine are direct consequences of its chemical structure and physical properties. The drug is most commonly supplied as a hydrochloride salt to improve its solubility for intravenous administration.[13] The distinction between the free base and the salt is critical; the inherent hydrophilicity of the molecule dictates its inability to passively diffuse across cell membranes, thereby necessitating the active nucleoside transport systems that are a key feature of its pharmacology.[4]
The structural and physical characteristics are summarized in Table 1.
Table 1: Drug Identification and Physicochemical Properties
Property | Value | Source(s) |
---|---|---|
IUPAC Name | 4-amino-1-pyrimidin-2-one | 13 |
Molecular Formula | C9H11F2N3O4 (free base) C9H11F2N3O4⋅HCl (hydrochloride salt) | 18 |
Molecular Weight | 263.20 g/mol (free base) 299.66 g/mol (hydrochloride salt) | 4 |
Physical Appearance | White to off-white crystalline solid/powder | 13 |
Melting Point | 168.64 °C (free base) 287-292 °C (decomposition, hydrochloride salt) | 13 |
Solubility | Hydrochloride Salt: Soluble in water, slightly soluble in methanol, practically insoluble in ethanol. Free Base (Research Grade): Often reported as soluble in DMSO, with limited water solubility. | 6 |
LogP (Octanol-Water Partition Coefficient) | -1.4 to -1.5 | 13 |
pKa (Dissociation Constant) | 3.6 | 13 |
SMILES | C1=CN(C(=O)N=C1N)[C@H]2C([C@@H]([C@H](O2)CO)O)(F)F | 13 |
InChIKey | SDUQYLNIPVEERB-QPPQHZFASA-N | 13 |
The clinical utility of Gemcitabine is rooted in its unique and complex pharmacology, encompassing its mechanism of action, its effects on the body (pharmacodynamics), and its absorption, distribution, metabolism, and excretion (pharmacokinetics).
Gemcitabine is a prodrug that exerts its cytotoxic effects after intracellular conversion to its active metabolites. Its mechanism is characterized by a requirement for active transport, a multi-step activation process, and a dual attack on DNA synthesis.[1]
Being a hydrophilic molecule, Gemcitabine cannot passively cross the lipid bilayer of cell membranes. Its entry into cancer cells is dependent on active transport mediated by protein carriers of the solute carrier (SLC) superfamily, specifically the human equilibrative nucleoside transporters (hENTs, e.g., SLC29A1) and human concentrative nucleoside transporters (hCNTs, e.g., SLC28A1, SLC28A3).[4]
Once inside the cell, Gemcitabine undergoes a critical three-step phosphorylation cascade to become pharmacologically active:
The activated metabolites of Gemcitabine, dFdCDP and dFdCTP, induce apoptosis (programmed cell death) through two distinct but interconnected mechanisms that create a powerful synergistic effect.[5]
This dual mechanism establishes an elegant "self-potentiating" pharmacological loop. The inhibition of RNR by dFdCDP lowers the concentration of the natural dCTP. This reduction in the competing natural substrate increases the likelihood that the fraudulent dFdCTP will be incorporated into DNA by DNA polymerase, thereby amplifying the drug's primary cytotoxic effect.[5] This sophisticated interplay is a key contributor to Gemcitabine's broad anti-tumor activity.
Due to its direct interference with DNA synthesis, Gemcitabine is a cell-cycle phase-specific agent. It primarily kills cells undergoing DNA synthesis in the S-phase and can also cause cell cycle arrest at the G1/S boundary.[1] Furthermore, Gemcitabine is a potent radiosensitizing agent. By depleting the deoxynucleotide pools required for DNA repair, it can enhance the cell-killing effects of radiation therapy.[1] This property is leveraged in combined-modality treatments for certain cancers.
The pharmacodynamic effects of Gemcitabine are characterized by schedule-dependent cytotoxicity and broad anti-tumor activity demonstrated in both preclinical models and clinical trials.
The antineoplastic effects of Gemcitabine are known to be schedule-dependent, meaning that the duration of exposure is a more critical determinant of efficacy than peak concentration. Prolonged infusions have been shown to enhance anti-tumor activity, a finding attributed to the saturation of the intracellular activating enzyme, DCK. However, this also increases toxicity, leading to the standard 30-minute infusion as a clinical compromise between efficacy and safety.[1]
Preclinically, Gemcitabine has demonstrated cytotoxic effects against a wide array of cancer cell lines and has inhibited the growth of human tumor xenografts from pancreatic, lung, breast, ovarian, and colon cancers by up to 99% in mouse models.[5]
In clinical settings, these effects translate to measurable patient outcomes. For example, in advanced NSCLC, monotherapy produced objective response rates (ORRs) of 18-26% with a median survival of 6-12 months, while combination with cisplatin yielded superior response rates. In advanced pancreatic cancer, a notoriously difficult-to-treat disease, Gemcitabine monotherapy established a benchmark with ORRs of 5-12% and a median survival of approximately 4-6 months.[5]
The absorption, distribution, metabolism, and excretion (ADME) profile of Gemcitabine is characterized by rapid clearance and a critical metabolic balance that dictates its activity and potential for resistance.
Gemcitabine is administered exclusively by intravenous infusion. Following a standard 30-minute infusion, peak plasma concentrations are reached within 15 to 30 minutes.[5] The volume of distribution is influenced by the infusion duration, increasing from approximately 50 L/m² for short infusions (<70 minutes) to 370 L/m² for longer infusions, indicating wider tissue distribution with prolonged exposure. Plasma protein binding is negligible at less than 10%. While the parent drug is cleared rapidly, the active triphosphate metabolite, dFdCTP, accumulates and is retained within tumor cells, where it exerts its prolonged cytotoxic effect.[5]
The metabolism of Gemcitabine represents a critical "tug-of-war" between activation and inactivation pathways, a balance that can determine clinical outcome and resistance.
This enzymatic balance has profound clinical implications. Low expression of the activating enzyme DCK or high expression of the inactivating enzyme CDA in a patient's tumor can lead to intrinsic or acquired resistance to Gemcitabine.[20] This pharmacogenomic variability suggests that patient response could potentially be predicted by measuring the expression levels of these enzymes and has spurred research into combination therapies with CDA inhibitors to enhance Gemcitabine's efficacy.[20]
Gemcitabine is eliminated predominantly through renal excretion. Within one week of administration, 92-98% of the dose is recovered in the urine, with over 89% being the inactive dFdU metabolite. Less than 10% of the dose is excreted as unchanged Gemcitabine.[5]
The terminal half-life of Gemcitabine is short and dependent on the infusion schedule, ranging from 32-94 minutes for short infusions to over 4 hours for longer infusions. In contrast, the active metabolite dFdCTP has a much longer intracellular half-life, ranging from 1.7 to 19.4 hours in mononuclear cells, which accounts for its sustained activity.[5] Systemic clearance is rapid, and studies have shown that clearance is approximately 30% lower in female patients and decreases with age, factors that may influence toxicity and require consideration during treatment.[5]
Gemcitabine is a broad-spectrum antineoplastic agent with established efficacy in a variety of solid tumors. Its role has evolved from a single-agent therapy to a foundational component of combination regimens, including recent integrations with immunotherapy that have expanded its therapeutic reach.
Gemcitabine is approved by the U.S. FDA and the EMA for the treatment of several advanced or metastatic cancers, primarily as part of a combination regimen. The specific approved indications and partner drugs are summarized in Table 2.
Table 2: Summary of Key FDA and EMA Approved Indications and Combination Regimens
Indication | Patient Population | Approved Combination Regimen | Regulatory Body (Key Approvals) |
---|---|---|---|
Pancreatic Cancer | Locally advanced or metastatic adenocarcinoma; first-line or after 5-FU failure | Gemcitabine Monotherapy | FDA (1996), EMA |
Non-Small Cell Lung Cancer (NSCLC) | Inoperable, locally advanced, or metastatic; first-line | Gemcitabine + Cisplatin | FDA (1998), EMA |
Metastatic Breast Cancer | First-line after failure of prior anthracycline-based chemotherapy | Gemcitabine + Paclitaxel | FDA (2004), EMA |
Advanced Ovarian Cancer | Relapsed disease ≥6 months after platinum-based therapy | Gemcitabine + Carboplatin | FDA, EMA |
Bladder Cancer | Locally advanced or metastatic | Gemcitabine + Cisplatin | EMA |
Biliary Tract Cancer (BTC) | Locally advanced unresectable or metastatic; first-line | Gemcitabine + Cisplatin + Pembrolizumab | FDA (2023) |
Urothelial Carcinoma (UC) | Unresectable or metastatic; first-line | Gemcitabine + Cisplatin + Nivolumab | FDA (2024) |
Beyond its approved indications, Gemcitabine has been extensively studied and is used off-label for a wide range of other malignancies. Completed Phase 2 clinical trials have demonstrated its activity in various lymphomas, including Hodgkin's lymphoma, mantle cell lymphoma, and cutaneous T-cell lymphoma, often in relapsed or refractory settings.[28] It is also a component of treatment regimens for mesothelioma and other solid tumors such as testicular cancer and sarcomas.[4]
The therapeutic landscape for Gemcitabine is undergoing a significant transformation, driven by its integration with novel agents and a deeper understanding of its biological effects.
A major recent development has been the successful combination of Gemcitabine with immune checkpoint inhibitors. This synergy is not merely additive; it is believed that Gemcitabine-induced immunogenic cell death primes the tumor microenvironment for a more robust immune response. By causing the release of tumor antigens and potentially depleting immunosuppressive cells like regulatory T cells (Tregs), Gemcitabine can "unmask" the tumor, making it more visible to the immune system. The subsequent administration of a checkpoint inhibitor then "releases the brakes" on T-cells, allowing them to mount an effective anti-tumor attack.[16]
This paradigm has led to major regulatory approvals:
The application of Gemcitabine is also moving beyond broad histological classifications toward more personalized, biomarker-driven strategies.
These developments underscore a strategic shift, leveraging Gemcitabine not just for its direct cytotoxicity but also for its ability to synergize with the most advanced therapies in oncology.
The safe and effective use of Gemcitabine requires strict adherence to established dosing regimens, schedules, and toxicity-driven dose modification guidelines. Administration is exclusively via intravenous infusion.
Dosages are calculated based on body surface area (BSA) and are typically administered as a 30-minute intravenous infusion. The specific dose and schedule vary significantly by indication and the combination agents used, as detailed in Table 3.[7] It is critical to note the FDA warning that infusion times prolonged beyond 60 minutes or dosing more frequently than once weekly are associated with a significant increase in toxicity, including hypotension and severe myelosuppression.[36]
Table 3: Recommended Dosing Regimens by Indication
Indication | Gemcitabine Dose (mg/m²) | Schedule | Combination Agent(s) and Dose |
---|---|---|---|
Ovarian Cancer | 1000 | Days 1 and 8 of a 21-day cycle | Carboplatin (AUC 4) on Day 1 after Gemcitabine |
Breast Cancer | 1250 | Days 1 and 8 of a 21-day cycle | Paclitaxel (175 mg/m²) on Day 1 before Gemcitabine |
NSCLC (4-week schedule) | 1000 | Days 1, 8, and 15 of a 28-day cycle | Cisplatin (100 mg/m²) on Day 1 after Gemcitabine |
NSCLC (3-week schedule) | 1250 | Days 1 and 8 of a 21-day cycle | Cisplatin (100 mg/m²) on Day 1 after Gemcitabine |
Pancreatic Cancer | 1000 | Weekly for 7 weeks, then 1 week rest. Subsequent cycles: weekly for 3 of every 4 weeks. | Monotherapy |
Dose modifications are an integral part of Gemcitabine therapy management, primarily driven by hematologic toxicity. The nadir for neutropenia typically occurs 7-10 days post-infusion, with recovery in the following week.[1] Dosing schedules and modification rules are designed around this kinetic profile to ensure patient safety. Complete blood counts, including differential and platelet counts, must be performed and assessed prior to each treatment cycle and on specified days within the cycle (e.g., Day 8).[35]
The specific thresholds for Absolute Neutrophil Count (ANC) and platelet count that trigger a dose reduction (e.g., to 75% or 50% of the full dose) or a delay in treatment vary by indication and are codified in the FDA-approved prescribing information. These guidelines represent a clinical risk-management algorithm based on the drug's predictable effect on the bone marrow. A summary of these guidelines for the most common indications is provided in Table 4.
Table 4: Selected Dose Modification Guidelines for Hematologic Toxicity on Day of Treatment
Indication | Treatment Day | Absolute Neutrophil Count (ANC) (x 10⁶/L) | Platelet Count (x 10⁶/L) | Required Dosage Modification |
---|---|---|---|---|
Ovarian Cancer | Day 1 | <1500 or | <100,000 | Delay Treatment Cycle |
Day 8 | 1000-1499 or | 75,000-99,999 | 50% of full dose | |
<1000 or | <75,000 | Hold | ||
Breast Cancer | Day 1 | <1500 or | <100,000 | Hold |
Day 8 | 1000-1199 or | 50,000-75,000 | 75% of full dose | |
700-999 and | ≥50,000 | 50% of full dose | ||
<700 or | <50,000 | Hold | ||
Pancreatic/NSCLC | Day of Tx | 500-999 or | 50,000-99,999 | 75% of full dose |
<500 or | <50,000 | Hold |
Note: This table is a simplified summary. Clinicians must refer to the full, most current prescribing information for complete details.[24]
Permanent dose reductions are warranted in cases of severe or prolonged myelosuppression, such as febrile neutropenia, an ANC below 500×106/L for more than 5 days, or a cycle delay exceeding one week due to toxicity.[7] For severe (Grade 3 or 4) non-hematologic toxicities, excluding nausea and vomiting, therapy should be held or the dose reduced by 50% based on physician judgment.[35]
Gemcitabine is supplied as either a lyophilized powder in single-dose vials requiring reconstitution or as a sterile, ready-to-use solution.[17] As a cytotoxic drug, it must be handled using appropriate safety precautions, including the use of protective gloves and following special disposal procedures.[40]
The calculated dose is withdrawn from the vial and typically diluted with 0.9% Sodium Chloride Injection to a final concentration of at least 0.1 mg/mL before administration.[39] The drug is classified as an irritant, and care must be taken to ensure proper venous access and to avoid extravasation, which can cause local tissue reaction.[1]
The clinical use of Gemcitabine is associated with a well-defined spectrum of adverse reactions, ranging from common, manageable side effects to rare but life-threatening toxicities. A thorough understanding of this safety profile is essential for patient monitoring and management.
The most common and clinically significant adverse reactions associated with Gemcitabine are detailed below and summarized in Table 5.
Adverse reactions occurring less frequently include diarrhea, constipation, stomatitis (mouth sores), alopecia (hair loss, usually mild), and infection.[1]
Table 5: Common and Serious Adverse Reactions by System Organ Class
System Organ Class | Adverse Reaction | Approximate Frequency | Clinical Notes/Management |
---|---|---|---|
Blood and Lymphatic System | Neutropenia, Anemia, Thrombocytopenia | Very Common (>60%) | Dose-limiting toxicity. Requires CBC monitoring before each dose. Dose modification based on ANC and platelet counts is critical. |
Febrile Neutropenia | Common | A serious complication requiring immediate medical attention and potential dose reduction. | |
Gastrointestinal | Nausea and Vomiting | Very Common (~69%) | Usually mild-moderate. Prophylactic antiemetics are recommended. |
Diarrhea, Stomatitis | Common | Supportive care, hydration. | |
Hepatic | Elevated Transaminases (ALT, AST) | Very Common | Usually transient and asymptomatic. Monitor liver function tests periodically. |
Constitutional | Flu-like Symptoms (Fever, Chills, Myalgia) | Very Common (~20-40%) | Usually mild and self-limiting. Acetaminophen may provide relief. |
Dermatologic | Rash, Pruritus | Very Common (~25%) | Typically mild. Topical corticosteroids or antihistamines may be used. |
Alopecia | Common | Hair loss is usually mild. | |
Respiratory | Dyspnea | Very Common | Usually mild. Unexplained or worsening dyspnea requires investigation for severe pulmonary toxicity. |
Pulmonary Toxicity (ARDS, Pneumonitis) | Rare | Potentially fatal. Discontinue Gemcitabine immediately. | |
Renal | Proteinuria, Hematuria | Very Common | Usually mild. Monitor renal function. |
Hemolytic Uremic Syndrome (HUS) | Rare | Potentially fatal. Discontinue Gemcitabine immediately. May not be reversible. | |
Cardiovascular | Edema (Peripheral, Generalized) | Very Common | Monitor for fluid retention. |
Capillary Leak Syndrome (CLS) | Rare | Potentially fatal. Discontinue Gemcitabine immediately. | |
Nervous System | Posterior Reversible Encephalopathy Syndrome (PRES) | Rare | Potentially fatal. Discontinue Gemcitabine immediately. |
Bolded items indicate clinically significant or life-threatening toxicities.
The FDA label for Gemcitabine includes several critical warnings and precautions.
A common pathological thread may link several of the most severe, non-hematologic toxicities. HUS/TMA, CLS, ARDS/pulmonary edema, and PRES can all be understood as clinical manifestations of widespread drug-induced endothelial cell injury. Damage to the small blood vessel lining can explain the micro-clotting and renal failure in HUS, the increased vascular permeability in CLS and pulmonary edema, and the breakdown of the blood-brain barrier in PRES.[1] This provides a unifying framework for recognizing these seemingly disparate but mechanistically related toxicities.
Gemcitabine has several clinically significant drug interactions, primarily related to its myelosuppressive and immunosuppressive effects.
Table 6: Clinically Significant Drug-Drug Interactions
Interacting Drug/Class | Potential Effect | Clinical Recommendation | Mechanism |
---|---|---|---|
Live Vaccines (e.g., BCG, MMR, Varicella) | Diminished vaccine efficacy and risk of disseminated infection. | Avoid. Live vaccines should be avoided for at least 3 months after therapy. | Pharmacodynamic antagonism due to immunosuppression. |
Other Immunosuppressants (e.g., tofacitinib, CAR-T therapies) | Additive immunosuppression, increased risk of severe infection. | Avoid or Use Alternate Drug. | Additive immunosuppressive effects. |
Cedazuridine | Increased Gemcitabine concentration and toxicity. | Avoid or Use Alternate Drug. | Inhibition of cytidine deaminase (CDA), the primary enzyme for Gemcitabine inactivation. |
Palifermin | Increased severity and duration of oral mucositis. | Avoid. Do not administer within 24 hours of Gemcitabine. | Increased toxicity, mechanism not fully elucidated. |
Warfarin | Increased anticoagulant effect (increased INR). | Use Caution/Monitor. | Potential for increased anticoagulant activity. |
Radiation Therapy | Severe and life-threatening radiation toxicity (e.g., mucositis, pneumonitis). | Avoid concurrent use. | Radiosensitization. |
Source(s): [4]
The trajectory of Gemcitabine from its initial synthesis to its current role as a mainstay of oncology provides a compelling case study in the lifecycle of a chemotherapeutic drug, marked by initial breakthroughs, broad expansion, and a modern renaissance through combination with novel therapies.
This evolution demonstrates how a deep understanding of a drug's mechanism, including its immunomodulatory effects, can unlock new therapeutic potential even decades after its initial approval.
Research into Gemcitabine remains highly active, focusing on overcoming its limitations and expanding its applications through innovative strategies.
Gemcitabine stands as a paradigm of a successful chemotherapeutic agent whose clinical value has not only endured but has been significantly enhanced over more than two decades of use. As a pyrimidine antimetabolite, its unique dual mechanism of action, involving both masked chain termination and self-potentiating inhibition of ribonucleotide reductase, provides a robust foundation for its broad-spectrum anti-tumor activity.
From its initial approval as a single-agent breakthrough for pancreatic cancer, Gemcitabine evolved into an essential backbone for combination chemotherapy regimens in lung, breast, ovarian, and bladder cancers. Its well-characterized and manageable toxicity profile, dominated by myelosuppression, has made it a reliable partner for a multitude of cytotoxic agents.
The contemporary era of oncology has ushered in a renaissance for Gemcitabine. Its integration into immunotherapy regimens has unlocked a new dimension of its pharmacology, leveraging its capacity to induce immunogenic cell death and prime the tumor microenvironment for attack by the host immune system. This has led to practice-changing approvals and has repositioned an established drug at the forefront of cancer treatment.
Future research focused on overcoming resistance, developing novel delivery systems, and refining its use through biomarker-driven personalization will continue to optimize the clinical application of this vital medication. Gemcitabine remains a testament to the enduring power of chemotherapy and its capacity for reinvention through intelligent and rational combination with the next generation of cancer therapies.
Published at: July 16, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.