Introduction: The Critical Role of EGFR in Cell Communication
Balance in the human body requires the ability of cells to communicate with each other and respond promptly to environmental cues. At the core of this communication lies signal transduction, where a signal produced by an external stimulus is propagated from the cell surface to the nucleus through a signaling cascade.
The receptor tyrosine kinase (RTKs) family is integral to the commencement of signal transduction, as it comprises receptors that can bind external stimuli. Among the RTKs, the epidermal growth factor receptor (EGFR) stands out for the myriad signal transduction pathways it influences. Its main binding partners are cytokines and growth factors, small proteins secreted into the extracellular matrix (ECM). EGF receptor signaling pathways regulate fundamental cell fate decisions, including proliferation, differentiation and apoptosis.¹ Simultaneously, abnormalities in EGFR signaling contribute to cancer and many other diseases.²
What is the EGF Receptor Signaling Pathway?
The EGF Receptor Signaling Pathway is a signal transduction pathway that delivers external growth signals to transcription factors in the nucleus, orchestrating gene expression for cellular processes.¹
EGFR belongs to the ErbB family, which contains four structurally related RTKs, such as ErbB1 (EGFR), ErbB2, ErbB3 and ErbB4, all encoded by the viral oncogene ErbB.³ Each ErbB receptor consists of a ligand-binding ectodomain (extracellular) region, a transmembrane domain, and an intracellular cytoplasmic domain that interacts with other ErbBs and signaling cascades.³ Furthermore, ErbBs, including EGFR, form homodimers or heterodimers upon stimulation by a binding partner.⁴
The binding partners of EGFR include:
- Epidermal Growth Factor (EGF): The most well-characterized EGFR ligand, responsible for epithelial tissue development¹
- Transforming Growth Factor Alpha (TGF-α): involved in epithelial development and regeneration⁵
- Amphiregulin: partaking in tissue repair and inflammation⁶
- Betacellulin: involved in pancreatic beta-cell proliferation⁷
- Heparin-binding EGF-like Growth Factor (HB-EGF) is essential in wound healing and cardiac development¹
- Epiregulin, implicated in inflammatory responses and tissue regeneration⁸
- Neuregulins (NRG1–NRG4), which contribute to nervous system development⁹
Collectively, these ligands bind EGFR, facilitating its dimerization and phosphorylation of the tyrosine residues in its intracellular domain.¹⁰
Mechanism of EGFR Activation
EGFR activation is initiated by the extracellular ligands, such as EGF or TGF-α, which bind to the EGFR extracellular domain. This binding induces a conformational change that facilitates dimerization by exposing the concealed dimerization sites when a monomeric EGFR is inactive.¹
EGFR can form a homodimer with another EGFR or a heterodimer with another ErbB.¹¹ In both cases, dimerization is necessary for activation at the intracellular tyrosine kinase domain.¹⁰ Dimerization is followed by autophosphorylation, in which two receptors phosphorylate each other's kinase domains in a process called trans-activation.¹²
EGFR phosphorylation occurs at specific tyrosine residues, converting them to docking platforms for adaptor proteins, which activate downstream signaling pathways. The growth factor receptor-bound protein 2 (GRB2) and Src Homology 2 domain-containing (Shc) transforming protein 1 are among the main binding partners of active EGFR.² These proteins can recruit Son of Sevenless (SOS), which activates Ras GTPase and initiates the following pathways:
- Mitogen-activated protein kinase (MAPK) signaling pathway, where the signal is propagated across RAS-RAF-MEK-ERK proteins, and active ERK translocates to the nucleus to regulate gene expression.¹³
- EGFR can also recruit phosphoinositide 3-kinase (PI3K), which activates protein kinase B (AKT). This, in turn, activates the mammalian target of rapamycin (mTOR), a protein that controls cell growth, proliferation, motility and survival.¹⁴
- EGFR can activate phospholipase C gamma (PLCγ) to promote protein kinase C (PKC) activation. This pathway is critical for calcium signaling and cytoskeletal rearrangements.¹⁵
- EGFR phosphorylates the signal transducer and activator of the transcription (STAT) family, such as STAT1 and STAT3, which mediate inflammation, immune response, proliferation and differentiation.¹⁶'¹⁷
EGFR Signal Regulation: Endocytosis, Trafficking and Degradation
The EGF receptor signaling pathways are tightly controlled by endocytosis, trafficking and degradation to maintain homeostasis and prevent aberrant signaling.¹⁸
The active EGFR-ligand complex is internalized through clathrin-dependent or independent endocytosis. The EGFR endosomes can be recycled back to the plasma membrane, where they resume signaling. On the other hand, it can get ubiquitinated and directed to the lysosome, which induces its degradation to attenuate signal transduction.¹⁹
Inside the cell, EGFR can also partake in crosstalks with the Wnt/β-catenin pathway, another signaling pathway critical in development and homeostasis. It can control gene expression by supporting β-catenin accumulation and nuclear translocation, establishing a balance between cell growth and differentiation.²⁰
Furthermore, EGFR can translocate to the nucleus to act as a transcription regulator by binding the promoter regions of genes, such as cyclin D1, to manage cell fate decisions.²¹
EGFR and Cytokine Signaling: Crosstalks in Inflammation and Cancer
There is a bidirectional relationship between cytokines and EGFR signaling pathways that governs essential processes like inflammation, repair and regeneration and promotes tumor development.²²
Research suggests that proinflammatory cytokines, such as interleukins IL-1 and IL-6 and tumor necrosis factor-alpha (TNF-α), could induce upregulation of EGFR expression.²² In addition, Cytokine receptors and EGFR could synergistically activate intracellular signaling pathways, although this convergence could enhance proliferation and survival in cancer cells. EGFR/JAK/STAT3 and IL6/STAT3 axes are such pathways that work together to over-activate the STAT signaling pathway.²³
In return, EGFR and downstream signaling pathways can amplify cytokine expression and secretion, engendering sustained signaling that underlies inflammatory diseases and cancer.²⁴ Many of the cytokines upregulated in cancer are associated with dysregulated immune checkpoints in immunosuppression. In contrast, upregulated chemokines act as chemoattractants that recruit neutrophils, tumor-associated macrophages (TAMs) and regulatory T cells.²⁵ Both cancer cells and TAMs continue to secrete cytokines that further activate EGFR-related pathways.²⁶ Thus, a pro-tumorigenic loop is created.
EGFR Signaling in Cancer and Disease
EGFR mutations, overexpression and overactivation are common hallmarks of various human cancers, driving uncontrolled proliferation, angiogenesis, metastasis and resistance to apoptosis.²
EGFR is overexpressed in many cancers, mainly in breast and colorectal tumors.¹² Furthermore, exon deletions and point mutations are often observed in non-small cell lung cancer (NSCLC) and glioblastoma, rendering EGFR constitutively active independently of ligand binding.¹²
EGFR's role in many cancers was uncovered through extensive research. Below are some examples:
- In colorectal cancer, EGFR overexpression is more common than mutations.²⁷
- EGFR is also frequently overexpressed in triple-negative breast cancer, with inhibitors displaying limited efficacy.²⁸
- EGFR mutations are prevalent among non-smokers in NSCLC. In particular, T790M mutations, MET amplification and histological transformation are correlated with poor prognosis and resistance to EGFR inhibitors, such as erlotinib and gefitinib.²⁹'³¹
- The type III EGFR mutation (EGFRvIII) is frequently observed in glioblastoma, where the mutant EGFR lacks the extracellular ligand-binding domain yet remains active. This type of glioblastoma has an aggressive phenotype associated with angiogenesis and therapy resistance.³²
EGFR overexpression and mutations generally lead to sustained activation of signal transduction pathways, downregulation of tumor suppressors and apoptotic pathways, crosstalk with other RTK-related pathways, immune evasion and metastasis.²
Applications in Drug Discovery and Diagnostics
The broad impact of EGFR on cancerous signaling pathways makes it an attractive target for drug development and cell therapy strategies.
To that end, tyrosine kinase inhibitors (such as gefitinib, erlotinib and Osimertinib) and monoclonal antibodies (such as cetuximab and panitumumab) were developed to inhibit EGFR.³³ These inhibitors block EGFR activation by interfering with EGFR ligand binding.³³ However, their success was hampered by the EGFR mutants that do not require ligand binding for activation.²⁹ Furthermore, the mutations in downstream pathway components, such as KRAS and BRAF, rendered EGFR inhibition ineffective.³⁴
Next-generation EGFR inhibitors that can bind EGFR mutants are under investigation to overcome these bottlenecks.³⁵ Furthermore, combination therapies co-targeting EGFR and downstream pathways, such as MAPK and PI3K/AKT, have the potential to inhibit the aberrant signal transduction systemically.³⁶'³⁷
Nevertheless, developing more robust EGFR therapies requires a thorough study of EGFR signaling in disease models. In vitro, in vivo and organoid models could reveal the mechanisms leading to EGFR mutation, drug resistance and heterogeneity in patient profiles, paving the way for more accurate patient stratification and personalized medicine.³⁸
Cellular assays and screening methods are central to developing accurate disease models. Cell-based assays and high-content imaging could be used to monitor EGFR dimerization and phosphorylation.³⁹ Furthermore, high-throughput screening can help identify small molecule inhibitors and guide the structural optimization of lead compounds, improving the chances of clinical success.⁴⁰
See how Danaher Life Sciences can help
FAQs
What happens to the EGF receptor after its ligand binds to it?
Ligand binding induces a conformational change in EGFR, leading to receptor dimerization (either homo- or heterodimers) and autophosphorylation of tyrosine residues in its cytoplasmic domain.²
Which intracellular signaling pathways are activated by EGFR?
EGFR activates several major pathways, including MAPK/ERK, PI3K/AKT, JAK/STAT and PLCγ, all of which regulate diverse cellular processes.²
What are the key proteins in the EGF receptor pathway?
Important adaptor and effector proteins include GRB2, SOS, Shc, PI3K, AKT, STATs and ERK, which transmit signals from the activated receptor.²
How does EGFR signaling regulate cell proliferation and survival?
EGFR signaling promotes cell cycle progression and prevents apoptosis by upregulating pro-growth genes and activating survival pathways like AKT.¹⁴
What is the role of cytokines in modulating the EGF receptor signaling pathway?
Cytokines can enhance or modulate EGFR signaling by inducing ligand shedding or interacting with EGFR pathways to amplify inflammation or tumor progression.⁴¹
References
- Tito C, Masciarelli S, Colotti G, Fazi F. EGF receptor in organ development, tissue homeostasis and regeneration. J Biomed Sci 2025;32(1):24.
- Levantini E, Maroni G, Del Re M, Tenen DG. EGFR signaling pathway as therapeutic target in human cancers. Semin Cancer Biol: Elsevier; 2022:253-275.
- Wu S, Haltom J, Zhao W, Yang J, Zhou Z, Gu X. Evolution and functional divergence of the ERBB receptor family. Pharmacogenomics 2021;22(8):473-484.
- Okada T, Miyagi H, Sako Y, Hiroshima M, Mochizuki A. Origin of diverse phosphorylation patterns in the ERBB system. Biophys J 2022;121(3):470-480.
- Murphrey MB, Quaim L, Rahimi N, Varacallo M. Biochemistry, epidermal growth factor receptor. StatPearls [Internet]: StatPearls Publishing; 2023.
- Singh SS, Chauhan SB, Kumar A, Kumar S, Engwerda CR, Sundar S, et al. Amphiregulin in cellular physiology, health, and disease: Potential use as a biomarker and therapeutic target. J Cell Physiol 2022;237(2):1143-1156.
- Lee Y-S, Song GJ, Jun H-S. Betacellulin-induced α-cell proliferation is mediated by ErbB3 and ErbB4, and may contribute to β-cell regeneration. Front Cell Dev Biol 2021;8:605110.
- Odell ID, Steach H, Gauld SB, Reinke-Breen L, Karman J, Carr TL, et al. Epiregulin is a dendritic cell–derived EGFR ligand that maintains skin and lung fibrosis. Sci Immunol 2022;7(78):eabq6691.
- Tagliaferro M, Ponti D. The signaling of neuregulin-epidermal growth factor receptors and its impact on the nervous system. Neuroglia 2023;4(4):253-274.
- Mudumbi KC, Burns EA, Schodt DJ, Petrova ZO, Kiyatkin A, Kim LW, et al. Distinct interactions stabilize EGFR dimers and higher-order oligomers in cell membranes. Cell Rep 2024;43(1).
- Bai X, Sun P, Wang X, Long C, Liao S, Dang S, et al. Structure and dynamics of the EGFR/HER2 heterodimer. Cell Discov 2023;9(1):18.
- Uribe ML, Marrocco I, Yarden Y. EGFR in cancer: Signaling mechanisms, drugs, and acquired resistance. Cancers (Basel) 2021;13(11):2748.
- Lin C-W, Nocka LM, Stinger BL, DeGrandchamp JB, Lew LN, Alvarez S, et al. A two-component protein condensate of the EGFR cytoplasmic tail and Grb2 regulates Ras activation by SOS at the membrane. PNAS 2022;119(19):e2122531119.
- Bang J, Jun M, Lee S, Moon H, Ro SW. Targeting EGFR/PI3K/AKT/mTOR signaling in hepatocellular carcinoma. Pharmaceutics 2023;15(8):2130.
- Huang W, Chen Q, Zhou P, Ye S, Fang Z. Neuroprotective effect of chrysophanol in Alzheimer disease via modulating the Ca2+/EGFR-PLCγ pathway. Neurosci Lett 2024;824:137684.
- Han Y, Zhang Y, Tian Y, Zhang M, Xiang C, Zhen Q, et al. The Interaction of the IFNγ/JAK/STAT1 and JAK/STAT3 Signalling Pathways in EGFR‐Mutated Lung Adenocarcinoma Cells. J Oncol 2022;2022(1):9016296.
- Hu X, Li J, Fu M, Zhao X, Wang W. The JAK/STAT signaling pathway: from bench to clinic. Signal Transduct Target Ther 2021;6(1):402.
- Schultz DF, Billadeau DD, Jois SD. EGFR trafficking: effect of dimerization, dynamics, and mutation. Front Oncol 2023;13:1258371.
- Zhou Y, Sakurai H. New trend in ligand-induced EGFR trafficking: A dual-mode clathrin-mediated endocytosis model. J Proteomics 2022;255:104503.
- Singh G, Hossain MM, Bhat AQ, Ayaz MO, Bano N, Eachkoti R, et al. Identification of a cross-talk between EGFR and Wnt/beta-catenin signaling pathways in HepG2 liver cancer cells. Cell Signal 2021;79:109885.
- Marijić B, Braut T, Babarović E, Krstulja M, Maržić D, Avirović M, et al. Nuclear EGFR expression is associated with poor survival in laryngeal carcinoma. Appl Immunohistochem Mol Morphol 2021;29(8):576-584.
- Shang D, Sun D, Shi C, Xu J, Shen M, Hu X, et al. Activation of epidermal growth factor receptor signaling mediates cellular senescence induced by certain pro‐inflammatory cytokines. Aging Cell 2020;19(5):e13145.
- Colyn L, Alvarez-Sola G, Latasa MU, Uriarte I, Herranz JM, Arechederra M, et al. New molecular mechanisms in cholangiocarcinoma: signals triggering interleukin-6 production in tumor cells and KRAS co-opted epigenetic mediators driving metabolic reprogramming. J Exp Clin Cancer Res 2022;41(1):183.
- Lu N, Wang L, Cao H, Liu L, Van Kaer L, Washington MK, et al. Activation of the epidermal growth factor receptor in macrophages regulates cytokine production and experimental colitis. J Immunol 2014;192(3):1013-1023.
- Propper DJ, Balkwill FR. Harnessing cytokines and chemokines for cancer therapy. Nat Rev Clin Oncol 2022;19(4):237-253.
- Wang X, Semba T, Manyam GC, Wang J, Shao S, Bertucci F, et al. EGFR is a master switch between immunosuppressive and immunoactive tumor microenvironment in inflammatory breast cancer. Sci Adv 2022;8(50):eabn7983.
- Janani B, Vijayakumar M, Priya K, Kim JH, Prabakaran D, Shahid M, et al. EGFR-Based targeted therapy for colorectal cancer—Promises and challenges. Vaccines 2022;10(4):499.
- Wu X, Sheng H, Zhao L, Jiang M, Lou H, Miao Y, et al. Co-loaded lapatinib/PAB by ferritin nanoparticles eliminated ECM-detached cluster cells via modulating EGFR in triple-negative breast cancer. Cell Death Dis 2022;13(6):557.
- Vaclova T, Grazini U, Ward L, O’Neill D, Markovets A, Huang X, et al. Clinical impact of subclonal EGFR T790M mutations in advanced-stage EGFR-mutant non-small-cell lung cancers. Nat Commun 2021;12(1):1780.
- Qin K, Hong L, Zhang J, Le X. MET amplification as a resistance driver to TKI therapies in lung cancer: clinical challenges and opportunities. Cancers (Basel) 2023;15(3):612.
- Pathak R, Villaflor VM. Histologic transformation in EGFR-mutant lung adenocarcinomas: mechanisms and therapeutic implications. Cancers (Basel) 2021;13(18):4641.
- Hoogstrate Y, Ghisai SA, de Wit M, de Heer I, Draaisma K, van Riet J, et al. The EGFRvIII transcriptome in glioblastoma: A meta-omics analysis. Neuro Oncol 2022;24(3):429-441.
- Zubair T, Bandyopadhyay D. Small Molecule EGFR Inhibitors as Anti-Cancer Agents: Discovery, Mechanisms of Action, and Opportunities. Int J Mol Sci 2023;24(3):2651.
- Dao V, Heestand G. Beyond EGFR inhibitors in advanced colorectal cancer: Targeting BRAF and HER2. Curr Probl Cancer 2023;47(4):100960.
- Li Y, Mao T, Wang J, Zheng H, Hu Z, Cao P, et al. Toward the next generation EGFR inhibitors: an overview of osimertinib resistance mediated by EGFR mutations in non-small cell lung cancer. Cell Commun Signal 2023;21(1):71.
- Tan L, Tran B, Tie J, Markman B, Ananda S, Tebbutt NC, et al. A phase Ib/II trial of combined BRAF and EGFR inhibition in BRAF V600E positive metastatic colorectal cancer and other cancers: the EVICT (erlotinib and vemurafenib in combination trial) study. Clin Cancer Res 2023;29(6):1017-1030.
- Ganesan K, Xu C, Wu J, Du B, Liu Q, Sui Y, et al. Ononin inhibits triple-negative breast cancer lung metastasis by targeting the EGFR-mediated PI3K/Akt/mTOR pathway. Sci China Life Sci 2024;67(9):1849-1866.
- Loewa A, Feng JJ, Hedtrich S. Human disease models in drug development. Nat Rev Bioeng 2023;1(8):545-559.
- Lei Q, Zhou X, Li Y, Zhao S, Yang N, Xiao Z, et al. Image-Based Phenotypic Profiling Enables Rapid and Accurate Assessment of EGFR-Activating Mutations in Tissues from Lung Cancer Patients. Journal of the American Chemical Society 2025.
- Robichaux JP, Le X, Vijayan R, Hicks JK, Heeke S, Elamin YY, et al. Structure-based classification predicts drug response in EGFR-mutant NSCLC. Nature 2021;597(7878):732-737.
- Mechelke T, Wittig F, Ramer R, Hinz B. Interleukin-1β induces tissue factor expression in A549 cells via EGFR-dependent and-independent mechanisms. Int J Mol Sci 2021;22(12):6606.
recent-articles