Gleevec, Glivec, Imatinib Teva, Imatinib Accord
Small Molecule
C29H31N7O
152459-95-5
Accelerated Phase Chronic Myelogenous Leukemia (CML), Aggressive Systemic Mastocytosis, Chordomas, Chronic Eosinophilic Leukemia, FIP1L1-PDGFRα fusion kinase status unknown Chronic eosinophilic leukemia, FIP1L1-PDGFRα fusion kinase status unknown Hypereosinophilic syndrome, Gastrointestinal Stromal Tumor (GIST), Hypereosinophilic Syndrome (HES), Metastatic Gastrointestinal Stromal Tumor (GIST), Metastatic Melanoma, Myelodysplastic Syndrome, Myeloproliferative Disorders (MPD), Refractory Acute Lymphoblastic Leukemia (ALL), Blast phase Chronic myeloid leukemia, CKit mutational status unknown Aggressive systemic mastocytosis, Metastatic Dermatofibrosarcoma protuberans, Newly diagnosed Acute Lymphoblastic Leukaemia, Newly diagnosed, chronic phase Chronic myeloid leukemia, Recurrent Dermatofibrosarcoma protuberans, Refractory, chronic phase Chronic myeloid leukemia, Unresectable Gastrointestinal stromal tumor
Imatinib is a small molecule kinase inhibitor that represents a watershed moment in the history of medicine and oncology. Its introduction in 2001 fundamentally altered the treatment of cancer, most notably Chronic Myeloid Leukemia (CML).[1] The drug's unprecedented clinical success led to it being hailed as a "miracle drug" and a "magical bullet," as it transformed CML from a rapidly fatal disease into a manageable, chronic condition for the majority of patients.[1]
Beyond its therapeutic impact, the development and success of imatinib established and validated a new paradigm in drug development known as "targeted therapy".[1] This approach moves away from the indiscriminate cytotoxicity of traditional chemotherapy and toward treatments tailored to the specific genetic and molecular abnormalities driving an individual patient's cancer.[1] Imatinib proved that by identifying a critical oncogenic driver and designing a drug to specifically inhibit it, remarkable efficacy could be achieved with a more manageable toxicity profile. This principle has since become the cornerstone of modern precision oncology, guiding the development of hundreds of subsequent targeted agents.
The story of imatinib is a testament to the power of rational drug design, built upon decades of fundamental scientific discovery. The narrative began in 1960 with the identification of an abnormally small chromosome 22 in patients with CML, an alteration that became known as the Philadelphia (Ph) chromosome.[5] Years later, this was understood to be the result of a reciprocal translocation between chromosomes 9 and 22, denoted as
t(9;22).[6] This breakthrough provided a specific, disease-defining genetic marker.
Subsequent research revealed that this translocation results in the molecular juxtaposition of the Breakpoint Cluster Region (BCR) gene on chromosome 22 and the Abelson murine leukemia viral oncogene homolog 1 (ABL1) gene on chromosome 9.[6] The product of this aberrant gene is a chimeric oncoprotein known as Bcr-Abl.[6] Critically, the Bcr-Abl protein is a constitutively active tyrosine kinase, meaning it is perpetually "switched on," driving uncontrolled cell proliferation and inhibiting apoptosis—the key pathogenetic events in CML.[6] The discovery of this single, causative molecular abnormality provided the ideal target for a rational drug discovery program.
In the early 1990s, a team at the pharmaceutical company Ciba-Geigy (a predecessor of Novartis), led by biochemist Nicholas Lyndon, began a concerted effort to screen for and develop a compound that could specifically inhibit the ATP-binding site of the Abl kinase.[3] After synthesizing and testing numerous 2-phenylaminopyrimidine derivatives, one candidate compound, STI-571, emerged as a potent and specific Bcr-Abl inhibitor.[5] The clinical development of this compound was championed by Dr. Brian Druker, an oncologist at the Dana-Farber Cancer Institute, who recognized its immense potential and drove it forward into human trials.[3] This linear, target-driven pathway—from identifying a genetic marker to understanding its protein product and then designing a drug to inhibit it—became the blueprint for a new generation of cancer therapies.
The first Phase I clinical trial of imatinib (then STI-571) commenced in June 1998.[5] The results were immediate and extraordinary. Between September 1998 and April 1999, investigators observed that nearly every CML patient treated with the drug experienced a significant and rapid response.[5] These remarkable findings, presented at the American Society of Hematology meeting in December 1999, galvanized the medical community and the manufacturer.[5]
Recognizing the drug's transformative potential, the U.S. Food and Drug Administration (FDA) granted it a fast-track designation, particularly for patients with advanced CML for whom few effective and well-tolerated treatments existed.[5] Novartis committed unprecedented resources to accelerate its development. Following the submission of a New Drug Application (NDA), the FDA approved Gleevec in a record-breaking 10 weeks, marking the fastest review period for any cancer drug at that time.[5] The initial FDA approval was granted on May 10, 2001, for the treatment of CML in blast crisis, accelerated phase, or in chronic phase after failure of interferon-alpha therapy.[3] This was quickly followed by approval from the European Medicines Agency (EMA) on November 7, 2001.[1] Over the subsequent years, the indications for imatinib were progressively expanded to include first-line CML treatment, various other leukemias, and solid tumors, with new formulations also gaining approval.[5]
Imatinib is a well-characterized small molecule drug with multiple identifiers used across scientific and regulatory databases. The clinically administered form is the mesylate salt, which has different properties and identifiers from the free base.
Imatinib is a synthetic organic compound classified as a 2-phenylaminopyrimidine derivative.[17] Its complex structure incorporates several key chemical moieties, including pyridine, pyrimidine, benzamide, and N-methylpiperazine groups, which contribute to its binding affinity and pharmacokinetic properties.[1] It is obtained by the formal condensation of the carboxy group of 4-[(4-methylpiperazin-1-yl)methyl]benzoic acid with the primary amino group of 4-methyl-N(3)-[4-(pyridin-3-yl)pyrimidin-2-yl]benzene-1,3-diamine.[19]
The decision to develop the mesylate salt was a critical step in creating a viable drug product. This salt form possesses superior physicochemical properties for oral administration compared to the free base.
Table 2.1: Key Chemical Identifiers and Properties of Imatinib | ||
---|---|---|
Property | Imatinib (Free Base) | Imatinib Mesylate |
DrugBank ID | DB00619 12 | DB00619 (active moiety) |
CAS Number | 152459-95-5 1 | 220127-57-1 13 |
Molecular Formula | C29H31N7O 1 | C29H31N7O⋅CH4SO3 8 |
Molecular Weight (Average) | 493.62 g/mol 20 | 589.71 g/mol 13 |
IUPAC Name | 4-[(4-methylpiperazin-1-yl)methyl]-N-[4-methyl-3-[(4-pyridin-3-ylpyrimidin-2-yl)amino]phenyl]benzamide 1 | N-(4-methyl-3-((4-(pyridin-3-yl)pyrimidin-2-yl)amino)phenyl)-4-((4-methylpiperazin-1-yl)methyl)benzamide methanesulfonate 13 |
SMILES | CC1=C(C=C(C=C1)NC(=O)C2=CC=C(C=C2)CN3CCN(CC3)C)NC4=NC=CC(=N4)C5=CN=CC=C5 1 | O=C(NC1=CC=C(C)C(NC2=NC=CC(C3=CC=CN=C3)=N2)=C1)C4=CC=C(CN5CCN(C)CC5)C=C4.CS(=O)(O)=O 13 |
InChIKey | KTUFNOKKBVMGRW-UHFFFAOYSA-N 1 | YLMAHDNUQAMNNX-UHFFFAOYSA-N 13 |
Key Solubilities | Poorly soluble in water | Very soluble in water; soluble in DMSO 8 |
Imatinib is classified as a Type-2 protein-tyrosine kinase inhibitor, a class of drugs that binds to the inactive conformation of the kinase domain.[7] It functions as a competitive inhibitor of adenosine triphosphate (ATP) by occupying the ATP-binding pocket of its target kinases.[17] This binding action prevents the enzyme from transferring a terminal phosphate group from ATP to tyrosine residues on its protein substrates (phosphorylation). By blocking this critical step, imatinib effectively shuts down the downstream signaling cascades that regulate fundamental cellular processes, including proliferation, survival, and differentiation, ultimately leading to cell death in cancer cells dependent on these pathways.[22]
The primary and most renowned target of imatinib is the Bcr-Abl tyrosine kinase, the constitutively active oncoprotein that is the hallmark of CML and a subset of Acute Lymphoblastic Leukemia (ALL).[6] Imatinib exhibits high potency and selectivity for Bcr-Abl.[7] It binds to a pocket on the Bcr-Abl kinase domain that is accessible only when the enzyme is in its inactive, non-functional conformation, thereby stabilizing this state and preventing its activation.[7] This blockade of Bcr-Abl activity halts the transmission of aberrant proliferative signals to the nucleus and induces apoptosis (programmed cell death) specifically in the malignant, Philadelphia chromosome-positive cells, while having minimal effect on normal, healthy cells that do not express this fusion protein.[6]
While designed to target Bcr-Abl, imatinib was discovered to be a potent inhibitor of several other key receptor tyrosine kinases. This broader activity profile is not a collection of unwanted side effects but is central to its efficacy in other malignancies. Imatinib potently inhibits the stem cell factor (SCF) receptor, known as c-KIT, and both alpha and beta platelet-derived growth factor receptors (PDGFR).[6]
This mechanism is the basis for its dramatic efficacy in Gastrointestinal Stromal Tumors (GIST), a majority of which are driven by gain-of-function mutations in the c−KIT gene that lead to its constitutive, ligand-independent activation.[6] Imatinib interrupts this aberrant KIT-mediated signaling, thereby inhibiting cell proliferation and inducing apoptosis in GIST cells.[6] Similarly, its activity against PDGFR explains its use in rare myelodysplastic/myeloproliferative diseases (MDS/MPD) that are characterized by genetic rearrangements involving the
PDGFR gene.[5] This demonstrates that the drug's utility is defined by its molecular targets rather than the specific anatomical site of the cancer.
The inhibitory spectrum of imatinib extends to other kinases, including the RET proto-oncogene, Macrophage colony-stimulating factor 1 receptor (CSF1R), and Epithelial discoidin domain-containing receptor 1 (DDR1).[12] This wider range of targets may contribute to both its therapeutic effects and its side effect profile. Furthermore, preclinical research has uncovered novel activities for imatinib, suggesting potential for drug repurposing. It has been reported to block the fusion of SARS-CoV and MERS-CoV with host cells and to enhance the host macrophage response to
Mycobacterium tuberculosis, indicating potential applications in infectious diseases.[10]
At the cellular level, the inhibition of Bcr-Abl, c-KIT, and PDGFR by imatinib disrupts a host of critical downstream signaling pathways that are hijacked by cancer cells. These include the Ras/MAPK pathway, which controls cellular proliferation; the Src/Pax/Fak/Rac pathway, which governs cellular motility and invasion; and the PI3K/AKT/BCL-2 pathway, a key regulator of apoptosis and cell survival.[12] By simultaneously blocking these pathways, imatinib delivers a multi-pronged attack on the cancer cell's core survival machinery.
The clinical pharmacodynamic effects are a direct reflection of this molecular action. In CML patients, a clear concentration-dependent decrease in white blood cell (WBC) counts is observed, providing a measurable indicator of drug activity.[32] A pharmacodynamic relationship has also been established between the steady-state plasma concentration of imatinib and the probability of developing edema, one of its most common side effects, highlighting the link between drug exposure and toxicity.[32]
The pharmacokinetic profile of imatinib is well-characterized and is highly favorable for a chronic oral therapy.
Imatinib is rapidly and almost completely absorbed following oral administration, with a mean absolute bioavailability of 98%.[8] This high and consistent bioavailability is a key feature, ensuring reliable drug exposure with oral dosing. Peak plasma concentrations (
Cmax) are typically reached within 2 to 4 hours after a dose.[8] While administration with a high-fat meal can slightly reduce the rate of absorption, it does not have a clinically significant effect on the overall extent of bioavailability.[29] It is recommended that imatinib be taken with a meal and a large glass of water to minimize the potential for gastrointestinal irritation.[26]
Once absorbed into the bloodstream, imatinib is extensively bound to plasma proteins (approximately 95%), primarily to albumin and α1-acid glycoprotein.[25] This high degree of protein binding influences its distribution and clearance. With once-daily dosing, the drug accumulates to a steady state, with plasma concentrations increasing by 1.5- to 2.5-fold compared to the first dose.[12]
Imatinib undergoes extensive metabolism, predominantly in the liver, via the cytochrome P450 (CYP) enzyme system.[6] The major enzyme responsible for its biotransformation is
CYP3A4.[6] Other CYP isoforms, including CYP1A2, CYP2D6, CYP2C9, and CYP2C19, play only a minor role in its metabolism.[6]
The primary metabolic pathway is N-demethylation of the piperazine moiety, which is mediated mainly by CYP3A4.[6] This process forms the main circulating metabolite, known as CGP 74588. This N-desmethyl derivative is pharmacologically active, with an
in vitro potency similar to that of the parent drug, imatinib.[6] However, its systemic exposure, as measured by the plasma area under the curve (AUC), is only about 15-16% of the parent drug's AUC, meaning imatinib itself is the primary contributor to the overall clinical effect.[8]
A critical aspect of imatinib's pharmacology is that it is not only a substrate of CYP3A4 but also a potent inhibitor of the enzyme.[32] Research has further defined it as a mechanism-based inhibitor, meaning it can irreversibly inactivate CYP3A4.[38] This dual role—where the drug inhibits its own primary clearance pathway—is a major contributor to the high inter-patient variability (around 40%) observed in its clearance and is the mechanistic basis for its extensive drug-drug interaction profile.[12]
Imatinib and its metabolites are eliminated from the body primarily through biliary excretion into the feces. Within 7 days of an oral dose, approximately 68% is recovered in the feces and 13% in the urine, mostly in the form of metabolites.[25] The elimination half-life (
t1/2) of the parent drug is approximately 18 hours, while the active metabolite (CGP 74588) has a longer half-life of about 40 hours.[7] The relatively long half-life of imatinib supports a convenient once-daily dosing schedule, which is beneficial for patient adherence in a chronic therapy setting.[7]
Table 3.1: Summary of Imatinib Pharmacokinetic Parameters | |
---|---|
Parameter | Value / Description |
Absolute Bioavailability | 98% 8 |
Time to Peak Concentration (Tmax) | 2–4 hours 8 |
Plasma Protein Binding | ~95% (mainly to albumin and α1-acid glycoprotein) 29 |
Primary Metabolic Enzyme | Cytochrome P450 3A4 (CYP3A4) 6 |
Active Metabolite | N-demethylated piperazine derivative (CGP 74588) 6 |
Elimination Half-life (t1/2) | Parent Drug: ~18 hours; Active Metabolite: ~40 hours 7 |
Primary Route of Elimination | Fecal excretion (~68% of dose), primarily as metabolites 25 |
Imatinib mesylate is approved by the U.S. FDA for a wide range of hematologic malignancies and solid tumors, reflecting its activity against multiple kinase targets. Its utility is defined by the presence of specific molecular markers rather than by tumor histology alone, a principle that underpins its broad application.
The approved indications include:
The clinical efficacy of imatinib is robustly supported by data from pivotal clinical trials that have redefined treatment standards.
The mechanism-based action of imatinib has led to its exploration in various other conditions where its molecular targets are implicated. Off-label use, where a physician prescribes a drug for an unapproved indication, is common in oncology, especially for targeted therapies when a patient's tumor expresses the relevant molecular target.[4]
The clinical journey of imatinib validates the "one drug, multiple targets, multiple diseases" concept. Its initial development was singularly focused on Bcr-Abl in CML, but its subsequent approvals for GIST (driven by c-KIT) and MDS/MPD (driven by PDGFR) demonstrate that a drug's true value is defined by its molecular interactions, not by the organ where the cancer originates. This principle has paved the way for modern "basket trials" and provides the scientific rationale for molecular profiling of tumors to identify patients who may benefit from a targeted drug, regardless of their cancer's histologic classification.
Table 4.1: FDA-Approved Indications for Imatinib with Recommended Dosing | ||
---|---|---|
Indication | Specific Patient Population | Recommended Daily Dose |
Ph+ CML (Chronic Phase) | Adults | 400 mg once daily 26 |
Ph+ CML (Accelerated Phase or Blast Crisis) | Adults | 600 mg once daily 26 |
Ph+ CML | Pediatrics | 340 mg/m²/day (once daily or split BID) 26 |
Ph+ ALL (Relapsed/Refractory) | Adults | 600 mg once daily 26 |
MDS/MPD | Adults with PDGFR gene rearrangements | 400 mg once daily 26 |
Aggressive Systemic Mastocytosis (ASM) | Adults without D816V c-Kit mutation | 100 mg or 400 mg once daily 26 |
HES/CEL | Adults with FIP1L1-PDGFRα fusion kinase | 100 mg or 400 mg once daily 26 |
Dermatofibrosarcoma Protuberans (DFSP) | Adults (unresectable, recurrent, or metastatic) | 800 mg (administered as 400 mg twice daily) 26 |
GIST (Metastatic and/or Unresectable) | Adults with Kit+ GIST | 400 mg once daily 26 |
GIST (Adjuvant Treatment) | Adults following resection of Kit+ GIST | 400 mg once daily 26 |
While imatinib is generally better tolerated than conventional cytotoxic chemotherapy, it is associated with a distinct and extensive profile of adverse reactions. These toxicities are largely a direct consequence of the inhibition of its target kinases (Abl, KIT, PDGFR) in healthy tissues where these pathways play normal physiological roles.
The most frequently reported adverse reactions, occurring in over 10% of patients, are generally low-grade and manageable. These include:
Other common side effects include headache, dizziness, and blurred vision, which may impact activities such as driving.[14]
Although less frequent, imatinib can cause severe and potentially life-threatening toxicities that require careful monitoring and management.
The FDA prescribing information for imatinib includes several important warnings and precautions, some of which are highlighted in a boxed warning.
The management of imatinib-related toxicities is a cornerstone of successful long-term therapy.
The safe use of imatinib requires careful consideration of its extensive potential for drug-drug and drug-food interactions. These interactions are a direct result of its metabolism by and inhibition of the cytochrome P450 enzyme system, making it a central hub for pharmacokinetic interactions.
Imatinib is primarily metabolized by CYP3A4, making its plasma concentration highly susceptible to modulation by other drugs that affect this enzyme.
Imatinib is not only a substrate but also an inhibitor of several key CYP enzymes. It is a moderate inhibitor of CYP3A4 and a potent inhibitor of CYP2D6 and CYP2C9.[1] This means imatinib can increase the plasma concentrations of other drugs that are metabolized by these enzymes, which is particularly dangerous for drugs with a narrow therapeutic index.
Patient education regarding food and supplement interactions is critical for safety.
A thorough medication reconciliation, including prescription drugs, over-the-counter products, and herbal supplements, is an absolute necessity before initiating and during imatinib therapy. The complexity of these interactions underscores the importance of a multidisciplinary care team involving physicians and pharmacists to ensure patient safety.
Table 6.1: Major Drug and Food Interactions with Imatinib | |||
---|---|---|---|
Interacting Agent/Class | Mechanism of Interaction | Clinical Consequence | Management Recommendation |
Strong CYP3A4 Inhibitors (e.g., Ketoconazole, Ritonavir, Clarithromycin) | Inhibition of imatinib metabolism | Increased imatinib plasma concentration; increased risk of toxicity | Avoid co-administration. If necessary, consider reducing imatinib dose with close monitoring. 8 |
Strong CYP3A4 Inducers (e.g., Rifampin, Phenytoin, Carbamazepine) | Induction of imatinib metabolism | Decreased imatinib plasma concentration; risk of therapeutic failure | Avoid co-administration. If necessary, a significant increase in imatinib dose may be required. 8 |
Grapefruit / Grapefruit Juice | Potent inhibition of intestinal CYP3A4 | Increased imatinib plasma concentration; increased risk of toxicity | Strictly avoid consumption during therapy. 45 |
St. John's Wort | Potent induction of CYP3A4 | Decreased imatinib plasma concentration; risk of therapeutic failure | Strictly avoid use during therapy. 44 |
CYP3A4 Substrates (e.g., Simvastatin, Amlodipine) | Imatinib inhibits CYP3A4 | Increased plasma concentration of the substrate drug; increased risk of its specific toxicities | Use with caution. Monitor for toxicity and consider dose reduction of the substrate drug or use of an alternative agent. 32 |
Warfarin | Imatinib inhibits CYP2C9, the primary metabolizing enzyme for warfarin | Increased warfarin levels; increased risk of bleeding | Avoid co-administration. Use alternative anticoagulants like low-molecular-weight heparin if possible. 45 |
Imatinib, marketed by Novartis, became a global pharmaceutical phenomenon under the brand names Gleevec in the United States and Glivec in Europe and other regions.[14] It rapidly achieved blockbuster status, with its revolutionary efficacy driving massive commercial success. In 2015, on the eve of its patent expiry in the US, global sales of Gleevec reached $4.66 billion.[52]
The patent expiration paved the way for generic competition. The first generic version of imatinib mesylate was launched in the United States by Sun Pharmaceuticals in February 2016, following FDA approval based on demonstrating bioequivalence to the branded product.[52] Since then, other manufacturers, such as Teva Pharmaceuticals, have also introduced generic versions, increasing market competition.[53] Even after generic entry, the market has seen the introduction of new branded formulations, such as Imkeldi, an oral solution approved in 2024, representing a strategy to serve specific patient populations and maintain a brand presence.[11]
The pricing of imatinib has been a focal point of the broader debate on the cost of cancer care. When it was first launched in 2001, the annual price was approximately $26,000.[52] However, over its patent life, the price increased dramatically. By 2016, the wholesale acquisition cost (WAC) for branded Gleevec in the US had soared to over $120,000 per year, and the average wholesale price (AWP) was over $145,000.[52]
The arrival of generics was anticipated to bring significant cost savings. However, the initial impact was muted. Due to market exclusivity provisions granted to the first generic filer under the Hatch-Waxman Act, the first generic from Sun Pharma was launched at a price only modestly lower than the branded drug, not the 30% discount that had been publicized.[52]
Despite the high price, numerous health economic analyses have been conducted. These studies consistently conclude that using lower-cost generic imatinib as the first-line therapy for CML is the most cost-effective strategy. This approach reserves the newer, more expensive second- and third-generation tyrosine kinase inhibitors (such as dasatinib, nilotinib, and ponatinib) for patients who fail or are intolerant to imatinib.[52] The economic lifecycle of imatinib—from a high-priced, paradigm-shifting branded drug to a more affordable generic standard of care—serves as a powerful case study on the tensions between rewarding pharmaceutical innovation, market dynamics, and the societal need for affordable access to life-saving medicines.
Imatinib will be remembered as one of the most important medical advances of the early 21st century. It was the agent that proved the principle of targeted cancer therapy, transforming a lethal cancer into a manageable chronic illness and giving hope and years of life to hundreds of thousands of patients worldwide. Its development story is a model of successful translational medicine, showcasing a seamless progression from fundamental biological discovery to rational drug design and definitive clinical validation.
The legacy of imatinib extends far beyond its direct therapeutic benefits. It fundamentally reshaped the landscape of oncology drug discovery, shifting the industry's focus toward molecularly targeted agents. It also ignited a global conversation about the value and cost of innovative medicines that continues to this day.
Looking forward, the story of imatinib is not over. Its role as a cost-effective, first-line therapy for CML and GIST is secure. Furthermore, ongoing research into its utility for non-oncologic conditions like multiple sclerosis and infectious diseases may yet uncover new applications for this remarkable molecule.[10] As cancer therapy moves toward complex combination regimens, imatinib will likely find new roles as a backbone agent. Its journey from a single-target "magic bullet" to a multi-purpose therapeutic tool with a complex economic and social history cements its place as a true landmark in medicine.
Published at: July 9, 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.