
Dacomitinib
Overview
Description
Dacomitinib is a medication primarily used for the treatment of non-small-cell lung carcinoma (NSCLC). It is marketed under the brand name Vizimpro. This compound is a selective and irreversible inhibitor of the epidermal growth factor receptor (EGFR) family, which includes EGFR/HER1, HER2, and HER4 . It was developed by Pfizer Inc. and approved by the FDA in September 2018 .
Preparation Methods
The preparation of dacomitinib involves several synthetic routes and reaction conditions. One method includes the following steps :
Methoxylation Reaction: N-(3-chloro-4-fluorophenyl)-7-fluoro-6-nitro-4-quinazolinamine is subjected to methoxylation in an alkali/methanol system to obtain N-(3-chloro-4-fluorophenyl)-7-methoxy-6-nitroquinazolinamine-4-amine.
Reduction Reaction: The compound obtained from the methoxylation reaction is then reduced using hydrazine hydrate to yield N-(3-chloro-4-fluorophenyl)-7-methoxyl-6-aminoquinazoline-4-amine.
Condensation Reaction: This compound is then condensed with an acyl chloride compound (2E)-4-(1-piperidyl)-2-butenoic acid hydrochloride in an N-methylpyrrolidone solvent to obtain a crude product.
Recrystallization: The crude product is refined in an ethanol-water solution to obtain this compound.
This method avoids the use of large amounts of solvent, making it more environmentally friendly and suitable for industrial production .
Chemical Reactions Analysis
Key Steps:
-
Hydrogenative Reduction :
Nitroarene 1 undergoes catalytic hydrogenation to form aniline 2 in acetonitrile. -
Amidation :
Aniline 2 reacts with butenoic acid 3 (prepared from bromide 6 ) using T3P (propane phosphonic acid anhydride) and 2,6-lutidine in acetonitrile. Reverse quenching with NaOH minimizes formamidine hydrolysis. -
Dimroth Rearrangement :
Intermediate 4 reacts with aniline 5 in neat acetic acid at 30°C to form dacomitinib. This step avoids high-temperature conditions typically required for Dimroth rearrangements .
Metabolic Reactions
This compound undergoes extensive phase I metabolism in rat liver microsomes (RLMs), primarily via piperidine ring hydroxylation and quinazoline ring reduction . Four major metabolites and two reactive intermediates were identified using LC-MS/MS :
Phase I Metabolites:
Metabolite | m/z | Fragment Ions | Retention Time (min) | Metabolic Pathway |
---|---|---|---|---|
DCB484 | 484 | 385, 319, 166, 144 | 39.5 | α-Oxidation at piperidine ring |
DCB472 | 472 | 387, 321, 152, 124 | 32.2 | Reduction at quinazoline ring |
DCB486a | 486 | 387, 166, 138 | 39.2 | Dual α-oxidation and reduction |
DCB486b | 486 | 385, 319, 168 | 30.4 | Hydroxylation at piperidine ring |
Bioactivation Pathways:
-
Iminium Ion Formation :
Piperidine ring oxidation generates iminium intermediates, stabilized as cyano adducts (DCB495, m/z 495) with KCN . -
Aldehyde Formation :
Oxidative dealkylation of the butenamide group produces an aldehyde intermediate, trapped as an oxime adduct (DCB362, m/z 362) with methoxylamine .
Enzyme-Mediated Reactions
This compound metabolism involves cytochrome P450 (CYP) enzymes :
-
CYP2D6 : Primary enzyme for O-desmethylation (PF-05199265, active metabolite).
-
CYP3A4 : Secondary pathway for oxidative metabolites.
Enzyme | Role | Metabolite |
---|---|---|
CYP2D6 | Oxidative O-desmethylation | PF-05199265 (active) |
CYP3A4 | Minor oxidation pathways | Uncharacterized |
Reactive Intermediates and Toxicity
Reactive intermediates formed during metabolism are linked to this compound’s adverse effects (e.g., hepatotoxicity) :
Intermediate | Structure | Trapping Agent | Adduct Detected | Toxicity Mechanism |
---|---|---|---|---|
Iminium ion | Piperidine-derived | KCN | DCB495 (cyano) | Protein/DNA alkylation |
Aldehyde | Butenamide-derived | Methoxylamine | DCB362 (oxime) | Glutathione depletion |
Elimination Pathways
Scientific Research Applications
First-Line Treatment for NSCLC
Dacomitinib has been approved as a first-line treatment for advanced NSCLC patients with classical EGFR mutations. The ARCHER 1050 trial demonstrated that this compound significantly improved progression-free survival (PFS) compared to gefitinib, establishing it as a standard option for this patient population .
Uncommon EGFR Mutations
Recent studies indicate that this compound is effective against NSCLC patients with uncommon EGFR mutations. A dual-center study found that patients with major uncommon mutations experienced favorable outcomes, suggesting this compound's potential beyond classical mutations .
Mutation Type | Response Rate | Study Reference |
---|---|---|
Classical Mutations | 70% stable disease | ARCHER 1050 trial |
Uncommon Mutations | Favorable activity | Dual-center cohort study |
Later-Line Treatment
This compound has shown efficacy in later-line settings for NSCLC patients who have progressed after prior therapies. A real-world study indicated that it remains well-tolerated and active in patients with various EGFR mutations, including those with brain metastases .
Recurrent Glioblastoma
This compound has also been investigated in glioblastoma patients with EGFR amplification. A Phase II trial evaluated its efficacy, revealing limited activity but some responses in specific cohorts, highlighting the need for further research into patient selection based on molecular characteristics .
Case Studies and Real-World Evidence
- Case Series on CNS Metastasis : A case series documented the effectiveness of this compound in treating CNS metastasis in NSCLC patients, demonstrating significant tumor reduction and manageable toxicity .
- Real-World Application : In a retrospective analysis involving over 100 patients treated with this compound after TKI resistance, the results showed a median PFS of approximately 4 months, indicating its potential as a subsequent therapy .
Safety Profile
This compound is generally well-tolerated, with manageable adverse effects such as diarrhea and rash being the most common. The incidence of severe adverse events is relatively low, making it a viable option for many patients.
Mechanism of Action
Dacomitinib exerts its effects by irreversibly inhibiting the kinase activity of the human EGFR family (EGFR/HER1, HER2, and HER4) and certain EGFR activating mutations (exon 19 deletion and exon 21 L858R substitution) . By binding to the ATP-binding site of these receptors, this compound prevents receptor autophosphorylation and downstream signaling, leading to the inhibition of cancer cell growth and proliferation .
Comparison with Similar Compounds
Dacomitinib is often compared to other EGFR inhibitors such as afatinib and gefitinib . Here are some key points of comparison:
Afatinib: Like this compound, afatinib is an irreversible EGFR inhibitor. Both drugs are used for the first-line treatment of EGFR mutation-positive NSCLC.
These comparisons highlight the uniqueness of this compound in terms of its irreversible binding and its efficacy in treating uncommon EGFR mutations.
Biological Activity
Dacomitinib is a second-generation, irreversible epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) primarily used in the treatment of non-small cell lung cancer (NSCLC) with specific EGFR mutations. Its mechanism of action involves covalent binding to the EGFR family members, including HER2 and HER4, leading to sustained inhibition of their kinase activity. This article aims to explore the biological activity of this compound, highlighting its efficacy, safety, and underlying mechanisms through various studies and clinical trials.
This compound irreversibly inhibits the tyrosine kinase activity of the EGFR family by forming a covalent bond with cysteine residues in the catalytic domain. This mechanism distinguishes it from reversible inhibitors, which compete for ATP binding without permanent modification. The potency of this compound has been demonstrated in various preclinical studies, showing an IC50 value as low as 6 nmol/L against wild-type and mutant EGFRs, including those with T790M resistance mutations .
Key Mechanisms:
- Covalent Bond Formation : this compound binds covalently to Cys773 in EGFR, preventing ATP binding and subsequent activation of downstream signaling pathways.
- Inhibition of Signaling Pathways : The blockade of EGFR signaling leads to reduced activation of critical pathways such as Ras-Raf-MAPK and PI3K/AKT, which are involved in cell proliferation and survival .
Phase II Trials
This compound has shown promising results in several clinical trials. For instance, the ARCHER 1050 trial demonstrated that this compound significantly improved progression-free survival (PFS) compared to gefitinib in patients with advanced NSCLC harboring sensitive EGFR mutations .
Summary of Clinical Findings:
- Objective Response Rate (ORR) : Approximately 70% in patients with EGFR mutations.
- Progression-Free Survival : Median PFS was reported at around 14.7 months for patients treated with this compound compared to 9.2 months for those on gefitinib .
- Overall Survival : The overall survival rate was also improved, although specific figures vary by study.
Case Studies
- Advanced NSCLC with Uncommon Mutations : A phase II trial investigated this compound's efficacy in patients with uncommon EGFR mutations (exons 18-21). Early results indicated that this compound could be effective in this subset, providing a new therapeutic option for patients who previously had limited treatment alternatives .
- Head and Neck Cancer : this compound has also been explored for its potential in treating squamous cell carcinoma of the head and neck (SCCHN). In preclinical models, it exhibited significant antitumor activity by downregulating HER signaling pathways and enhancing apoptosis through various mechanisms .
Safety Profile
The safety profile of this compound has been assessed across multiple studies. Common adverse effects include:
- Diarrhea
- Rash
- Fatigue
- Decreased appetite
Most side effects were manageable with dose adjustments or supportive care measures. Notably, only a small percentage (5%) of participants discontinued treatment due to adverse events .
Table: Summary of Adverse Effects
Adverse Effect | Incidence (%) | Management Strategies |
---|---|---|
Diarrhea | 60 | Loperamide or dose reduction |
Rash | 50 | Topical corticosteroids |
Fatigue | 40 | Supportive care |
Decreased Appetite | 30 | Nutritional support |
Q & A
Basic Research Questions
Q. What are the primary mechanisms of action of dacomitinib in EGFR-mutated non-small cell lung cancer (NSCLC), and how do they differ from first-generation EGFR inhibitors?
- Methodological Answer: this compound irreversibly inhibits pan-ErbB receptors (EGFR, HER2, HER4) by covalently binding to cysteine residues in the kinase domain, preventing ATP binding and downstream signaling. Unlike first-generation inhibitors (e.g., gefitinib), its irreversible binding enhances target suppression. Validate mechanisms using kinase assays (e.g., MTS assays for cell viability) and Western blotting to assess phosphorylation levels of EGFR and downstream effectors (e.g., AKT, MAPK) .
Q. How should researchers design in vitro studies to evaluate this compound resistance mechanisms?
- Methodological Answer: Use dose-escalation protocols in EGFR-mutated cell lines (e.g., PC9, H1975) over 6–12 months to simulate acquired resistance. Combine RNA sequencing and CRISPR-Cas9 screens to identify resistance-associated genes (e.g., MET amplification, EMT markers). Validate findings with colony formation assays and synergy studies with secondary inhibitors (e.g., MET inhibitors) .
Advanced Research Questions
Q. What are the critical considerations for designing clinical trials evaluating this compound in patients with hepatic impairment?
- Methodological Answer:
- Population: Stratify patients by Child-Pugh classification (A/B/C) and match with healthy controls.
- PK Analysis: Collect serial blood samples up to 144 hours post-dose. Use HPLC-MS/MS for plasma concentration analysis, ensuring a lower limit of quantification (LLOQ) ≤0.1 ng/mL.
- Statistical Approach: Apply non-compartmental analysis (NCA) via openNCA software to calculate AUC, Cmax, and t1/2. Account for protein binding using rapid equilibrium dialysis (RED) .
Q. How can researchers resolve contradictions in efficacy data between this compound monotherapy and combination regimens (e.g., with radiotherapy)?
- Methodological Answer: Conduct normalized isobologram analysis (Chou-Talalay method) to quantify synergy. In SCCHN models, combine this compound (5–100 nM) with ionizing radiation (2–4 Gy). Use clonogenic survival assays and measure apoptotic markers (e.g., caspase-3 cleavage). Address variability by standardizing irradiation protocols and cell line selection (e.g., FaDu vs. UT-SCC lines) .
Q. What methodologies are recommended for assessing this compound’s impact on tumor microenvironment (TME) dynamics?
- Methodological Answer: Utilize single-cell RNA sequencing (scRNA-seq) of patient-derived xenografts (PDXs) pre- and post-treatment. Focus on immune cell infiltration (e.g., CD8+ T cells, macrophages) and cytokine profiling (e.g., IL-6, TGF-β). Validate with multiplex immunohistochemistry (IHC) and spatial transcriptomics .
Q. How should pharmacokinetic/pharmacodynamic (PK/PD) models be optimized for this compound in pediatric populations?
- Methodological Answer: Develop physiologically based pharmacokinetic (PBPK) models incorporating age-related changes in CYP3A4 activity and plasma protein binding. Validate with sparse sampling in phase I trials, leveraging Bayesian estimation for AUC prediction. Prioritize safety endpoints (e.g., rash, diarrhea) due to higher toxicity risks .
Q. Methodological and Ethical Considerations
Q. What are the best practices for ensuring reproducibility in this compound pre-clinical studies?
- Methodological Answer:
- Experimental Replicates: Use ≥3 biological replicates per condition.
- Data Transparency: Publish raw HPLC-MS/MS chromatograms and RED assay validation data as supplementary materials.
- Reference Standards: Calibrate assays with this compound and metabolite (PF-05199265) reference standards .
Q. How can researchers address ethical challenges in trials involving this compound and vulnerable populations (e.g., severe hepatic impairment)?
- Methodological Answer: Exclude patients with hepatocellular carcinoma or hepatorenal syndrome to minimize confounding risks. Obtain written informed consent with explicit disclosure of off-label use risks. Include independent data monitoring committees (IDMCs) for interim safety reviews .
Q. Data Analysis and Reporting
Q. What statistical methods are robust for analyzing this compound’s long-term survival benefits in ARCHER 1050-like trials?
- Methodological Answer: Apply Cox proportional hazards models with stratification by EGFR mutation subtype (e.g., exon 19 del vs. L858R). Use landmark analysis to mitigate immortal time bias. Report median progression-free survival (PFS) with 95% confidence intervals .
Q. How should contradictory findings in this compound’s CNS penetration data be addressed?
- Methodological Answer: Perform cerebrospinal fluid (CSF) sampling in NSCLC patients with brain metastases. Compare CSF-to-plasma ratios using tandem mass spectrometry. Adjust for blood-brain barrier integrity via contrast-enhanced MRI findings .
Properties
IUPAC Name |
(E)-N-[4-(3-chloro-4-fluoroanilino)-7-methoxyquinazolin-6-yl]-4-piperidin-1-ylbut-2-enamide | |
---|---|---|
Source | PubChem | |
URL | https://pubchem.ncbi.nlm.nih.gov | |
Description | Data deposited in or computed by PubChem | |
InChI |
InChI=1S/C24H25ClFN5O2/c1-33-22-14-20-17(24(28-15-27-20)29-16-7-8-19(26)18(25)12-16)13-21(22)30-23(32)6-5-11-31-9-3-2-4-10-31/h5-8,12-15H,2-4,9-11H2,1H3,(H,30,32)(H,27,28,29)/b6-5+ | |
Source | PubChem | |
URL | https://pubchem.ncbi.nlm.nih.gov | |
Description | Data deposited in or computed by PubChem | |
InChI Key |
LVXJQMNHJWSHET-AATRIKPKSA-N | |
Source | PubChem | |
URL | https://pubchem.ncbi.nlm.nih.gov | |
Description | Data deposited in or computed by PubChem | |
Canonical SMILES |
COC1=C(C=C2C(=C1)N=CN=C2NC3=CC(=C(C=C3)F)Cl)NC(=O)C=CCN4CCCCC4 | |
Source | PubChem | |
URL | https://pubchem.ncbi.nlm.nih.gov | |
Description | Data deposited in or computed by PubChem | |
Isomeric SMILES |
COC1=C(C=C2C(=C1)N=CN=C2NC3=CC(=C(C=C3)F)Cl)NC(=O)/C=C/CN4CCCCC4 | |
Source | PubChem | |
URL | https://pubchem.ncbi.nlm.nih.gov | |
Description | Data deposited in or computed by PubChem | |
Molecular Formula |
C24H25ClFN5O2 | |
Source | PubChem | |
URL | https://pubchem.ncbi.nlm.nih.gov | |
Description | Data deposited in or computed by PubChem | |
DSSTOX Substance ID |
DTXSID50149493 | |
Record name | Dacomitinib | |
Source | EPA DSSTox | |
URL | https://comptox.epa.gov/dashboard/DTXSID50149493 | |
Description | DSSTox provides a high quality public chemistry resource for supporting improved predictive toxicology. | |
Molecular Weight |
469.9 g/mol | |
Source | PubChem | |
URL | https://pubchem.ncbi.nlm.nih.gov | |
Description | Data deposited in or computed by PubChem | |
Solubility |
<1 mg/mL | |
Record name | Dacomitinib | |
Source | DrugBank | |
URL | https://www.drugbank.ca/drugs/DB11963 | |
Description | The DrugBank database is a unique bioinformatics and cheminformatics resource that combines detailed drug (i.e. chemical, pharmacological and pharmaceutical) data with comprehensive drug target (i.e. sequence, structure, and pathway) information. | |
Explanation | Creative Common's Attribution-NonCommercial 4.0 International License (http://creativecommons.org/licenses/by-nc/4.0/legalcode) | |
Mechanism of Action |
Dacomitinib is an irreversible small molecule inhibitor of the activity of the human epidermal growth factor receptor (EGFR) family (EGFR/HER1, HER2, and HER4) tyrosine kinases. It achieves irreversible inhibition via covalent bonding to the cysteine residues in the catalytic domains of the HER receptors. The affinity of dacomitinib has been shown to have an IC50 of 6 nmol/L. The ErbB or epidermal growth factor (EGF) family plays a role in tumor growth, metastasis, and treatment resistance by activating downstream signal transduction pathways such as such as Ras-Raf-MAPK, PLCgamma-PKC-NFkB and PI3K/AKT through the tyrosine kinase-driven phosphorylation at the carboxy-terminus. Around 40% of cases show amplification of EGFR gene and 50% of the cases present the _EGFRvIII_ mutation which represents a deletion that produces a continuous activation of the tyrosine kinase domain of the receptor. | |
Record name | Dacomitinib | |
Source | DrugBank | |
URL | https://www.drugbank.ca/drugs/DB11963 | |
Description | The DrugBank database is a unique bioinformatics and cheminformatics resource that combines detailed drug (i.e. chemical, pharmacological and pharmaceutical) data with comprehensive drug target (i.e. sequence, structure, and pathway) information. | |
Explanation | Creative Common's Attribution-NonCommercial 4.0 International License (http://creativecommons.org/licenses/by-nc/4.0/legalcode) | |
CAS No. |
1110813-31-4 | |
Record name | Dacomitinib | |
Source | CAS Common Chemistry | |
URL | https://commonchemistry.cas.org/detail?cas_rn=1110813-31-4 | |
Description | CAS Common Chemistry is an open community resource for accessing chemical information. Nearly 500,000 chemical substances from CAS REGISTRY cover areas of community interest, including common and frequently regulated chemicals, and those relevant to high school and undergraduate chemistry classes. This chemical information, curated by our expert scientists, is provided in alignment with our mission as a division of the American Chemical Society. | |
Explanation | The data from CAS Common Chemistry is provided under a CC-BY-NC 4.0 license, unless otherwise stated. | |
Record name | Dacomitinib [USAN:INN] | |
Source | ChemIDplus | |
URL | https://pubchem.ncbi.nlm.nih.gov/substance/?source=chemidplus&sourceid=1110813314 | |
Description | ChemIDplus is a free, web search system that provides access to the structure and nomenclature authority files used for the identification of chemical substances cited in National Library of Medicine (NLM) databases, including the TOXNET system. | |
Record name | Dacomitinib | |
Source | DrugBank | |
URL | https://www.drugbank.ca/drugs/DB11963 | |
Description | The DrugBank database is a unique bioinformatics and cheminformatics resource that combines detailed drug (i.e. chemical, pharmacological and pharmaceutical) data with comprehensive drug target (i.e. sequence, structure, and pathway) information. | |
Explanation | Creative Common's Attribution-NonCommercial 4.0 International License (http://creativecommons.org/licenses/by-nc/4.0/legalcode) | |
Record name | Dacomitinib | |
Source | EPA DSSTox | |
URL | https://comptox.epa.gov/dashboard/DTXSID50149493 | |
Description | DSSTox provides a high quality public chemistry resource for supporting improved predictive toxicology. | |
Record name | DACOMITINIB ANHYDROUS | |
Source | FDA Global Substance Registration System (GSRS) | |
URL | https://gsrs.ncats.nih.gov/ginas/app/beta/substances/2XJX250C20 | |
Description | The FDA Global Substance Registration System (GSRS) enables the efficient and accurate exchange of information on what substances are in regulated products. Instead of relying on names, which vary across regulatory domains, countries, and regions, the GSRS knowledge base makes it possible for substances to be defined by standardized, scientific descriptions. | |
Explanation | Unless otherwise noted, the contents of the FDA website (www.fda.gov), both text and graphics, are not copyrighted. They are in the public domain and may be republished, reprinted and otherwise used freely by anyone without the need to obtain permission from FDA. Credit to the U.S. Food and Drug Administration as the source is appreciated but not required. | |
Melting Point |
184-187 ºC | |
Record name | Dacomitinib | |
Source | DrugBank | |
URL | https://www.drugbank.ca/drugs/DB11963 | |
Description | The DrugBank database is a unique bioinformatics and cheminformatics resource that combines detailed drug (i.e. chemical, pharmacological and pharmaceutical) data with comprehensive drug target (i.e. sequence, structure, and pathway) information. | |
Explanation | Creative Common's Attribution-NonCommercial 4.0 International License (http://creativecommons.org/licenses/by-nc/4.0/legalcode) | |
Retrosynthesis Analysis
AI-Powered Synthesis Planning: Our tool employs the Template_relevance Pistachio, Template_relevance Bkms_metabolic, Template_relevance Pistachio_ringbreaker, Template_relevance Reaxys, Template_relevance Reaxys_biocatalysis model, leveraging a vast database of chemical reactions to predict feasible synthetic routes.
One-Step Synthesis Focus: Specifically designed for one-step synthesis, it provides concise and direct routes for your target compounds, streamlining the synthesis process.
Accurate Predictions: Utilizing the extensive PISTACHIO, BKMS_METABOLIC, PISTACHIO_RINGBREAKER, REAXYS, REAXYS_BIOCATALYSIS database, our tool offers high-accuracy predictions, reflecting the latest in chemical research and data.
Strategy Settings
Precursor scoring | Relevance Heuristic |
---|---|
Min. plausibility | 0.01 |
Model | Template_relevance |
Template Set | Pistachio/Bkms_metabolic/Pistachio_ringbreaker/Reaxys/Reaxys_biocatalysis |
Top-N result to add to graph | 6 |
Feasible Synthetic Routes
Disclaimer and Information on In-Vitro Research Products
Please be aware that all articles and product information presented on BenchChem are intended solely for informational purposes. The products available for purchase on BenchChem are specifically designed for in-vitro studies, which are conducted outside of living organisms. In-vitro studies, derived from the Latin term "in glass," involve experiments performed in controlled laboratory settings using cells or tissues. It is important to note that these products are not categorized as medicines or drugs, and they have not received approval from the FDA for the prevention, treatment, or cure of any medical condition, ailment, or disease. We must emphasize that any form of bodily introduction of these products into humans or animals is strictly prohibited by law. It is essential to adhere to these guidelines to ensure compliance with legal and ethical standards in research and experimentation.