Epidermal Growth Factor-like 6 (EGFL6) is a secreted glycoprotein belonging to the epidermal growth factor (EGF) repeat superfamily. It is encoded by the EGFL6 gene located on the Xp22.2 chromosome in humans and plays critical roles in angiogenesis, tissue remodeling, and tumor progression . Unlike other EGF-like proteins, EGFL6 is highly expressed in tumor-associated endothelial and epithelial cells but shows minimal expression in normal adult tissues . Its dysregulation is strongly associated with aggressive cancers, including high-grade serous ovarian cancer (HGSOC), colorectal cancer, and breast cancer .
EGFL6 promotes endothelial cell migration, proliferation, and tube formation via:
EGFL6 induces myeloid cell differentiation into immunosuppressive phenotypes (e.g., MDSCs, TAMs) via Syk/ERK phosphorylation, reducing cytotoxic CD8+ T-cell infiltration .
Ovarian Cancer:
Therapeutic Targeting:
Prognostic Marker: Elevated EGFL6 correlates with poor survival in HGSOC (HR = 2.1, p < 0.01) .
Therapeutic Potential: Humanized antibodies targeting EGFL6 show promise in Phase I readiness, with no toxicity observed in murine models .
EGFL6 is a secreted protein belonging to the EGF-repeat superfamily that functions as a matricellular protein. In human tissues, EGFL6 shows a highly specific expression pattern with the highest expression observed in placenta, adipose tissue, and hypothalamus . This restricted expression profile in adult tissues makes it particularly interesting as a potential therapeutic target with potentially limited off-target effects. Importantly, while expression is restricted in normal adult tissues, EGFL6 becomes upregulated in various pathological states, particularly in malignancies and adipose tissue dysfunction.
EGFL6 expression is significantly higher in isolated adipocytes compared to stromal vascular fraction (SVF) cells. Quantitative analysis has shown that both adipocyte and SVF EGFL6 gene expression are significantly increased in children with obesity, with adipocyte expression showing a more pronounced increase (2.2-fold) compared to SVF cells (1.4-fold) . This differential expression suggests cell-specific regulation and function of EGFL6 within the adipose tissue microenvironment, pointing to adipocytes as the primary source of EGFL6 in adipose tissue dysfunction associated with obesity.
EGFL6 activates several key signaling pathways, with the ERK pathway being one of the most well-characterized. The protein stimulates ERK phosphorylation in responsive cancer cells . This activation of the ERK pathway contributes to multiple cellular processes that promote cancer progression, including increased cell migration, enhanced proliferation, and resistance to apoptosis. Mechanistically, EGFL6 likely functions through receptor binding on the cell surface, triggering downstream signaling cascades that ultimately result in ERK activation and subsequent cellular responses.
For robust EGFL6 detection, researchers should employ multiple complementary approaches. Western blotting can detect denatured EGFL6 protein, appearing as a band between 51-64 kDa markers in EGFL6-expressing samples . Specificity can be confirmed through blocking experiments using recombinant human EGFL6 protein. For detection of native EGFL6, immunofluorescence staining can be employed on cultured cells and tissue sections. Notably, anti-EGFL6 antibodies have successfully detected EGFL6 in both blood vessels and tumor cells in xenograft models . For quantitative analysis, ELISA can be used to measure EGFL6 levels in biological fluids and cell culture supernatants.
Several functional assays have been validated for assessing EGFL6 activity:
Migration assay: A wound healing/scratch assay can evaluate EGFL6-driven cancer cell migration. Treatment with recombinant EGFL6 protein increases "wound healing," which can be quantifiably inhibited by neutralizing antibodies .
Proliferation assay: Tumor sphere formation assays with cancer cell lines (both EGFL6-expressing and non-expressing) can measure EGFL6-induced proliferation effects .
Signaling activation: Western blotting for phosphorylated ERK (pERK) provides a direct measurement of EGFL6 signaling activity, with increased pERK levels observed after EGFL6 treatment .
These complementary approaches provide a comprehensive assessment of EGFL6's biological activities and can be used to evaluate the efficacy of potential EGFL6-targeting therapeutic agents.
Patient-derived xenograft (PDX) models incorporating cancer-associated mesenchymal stem cells (CA-MSCs) represent a stringent and clinically relevant system for evaluating EGFL6 function and therapeutic targeting. This approach offers several advantages:
PDX models maintain human tumor heterogeneity and microenvironment.
Incorporated CA-MSCs differentiate to create human cancer-associated fibroblasts.
These models enhance cancer growth, metastasis, and therapeutic resistance, better mimicking human disease .
For therapeutic evaluation, intraperitoneal administration of anti-EGFL6 antibodies (10 mg/kg biweekly) has proven effective in reducing tumor growth and preventing metastasis . Pharmacokinetic studies in mice expressing human FC receptors provide more accurate translation to human applications.
EGFL6 is highly expressed in high-grade serous ovarian cancer and serves as a biomarker of poor prognosis . This protein plays a dual role in promoting cancer progression by:
Stimulating endothelial cell proliferation and angiogenesis, enhancing blood supply to tumors.
Directly promoting cancer cell proliferation and metastasis through activation of signaling pathways including ERK phosphorylation .
The secreted nature of EGFL6 enables it to act in both autocrine and paracrine manners, influencing both cancer cells and the surrounding tumor microenvironment. Its relatively restricted expression in normal adult tissues makes it an attractive therapeutic target with potentially limited off-target effects.
Humanized affinity-matured anti-EGFL6 antibodies have demonstrated significant anti-tumor effects in preclinical models:
Parameter | Control IgG | Antibody #9 | Antibody #16 |
---|---|---|---|
Tumor growth reduction | - | 45.2% | 64.7% |
Metastasis incidence | 75% | 0% | 0% |
Major toxicities | None | None | None |
These antibodies function by neutralizing EGFL6, thereby inhibiting multiple processes critical for cancer progression:
Reduction in angiogenesis
Decreased tumor cell proliferation (reduced Ki67 expression)
The efficacy of these antibodies in stringent PDX models incorporating human cancer-associated mesenchymal stem cells supports their potential clinical utility.
The development of clinically viable anti-EGFL6 antibodies faces several methodological challenges:
Immunogenicity concerns: Murine antibodies can elicit significant immune responses in humans, necessitating humanization through complementarity determining region (CDR) grafting methods .
Affinity optimization: Even after humanization, antibody affinity must be optimized through techniques such as the FASEBA (Fast Screening for Expression, Biophysical-properties and Affinity) platform and NNK library screening .
Binding specificity verification: Multiple assays are needed to confirm specificity:
Functional neutralization testing: Candidate antibodies must be screened through multiple functional assays (migration, proliferation, ERK phosphorylation) to confirm neutralizing activity .
EGFL6 expression shows a strong positive association with adipocyte hypertrophy in obese individuals . This relationship extends to several parameters of metabolic dysfunction:
Higher EGFL6 expression correlates with increased adipocyte size, a hallmark of adipose tissue dysfunction.
EGFL6 expression is significantly elevated in adipose tissue of children with obesity.
The increased expression in obesity is observed in both adipocytes (2.2-fold) and stromal vascular fraction cells (1.4-fold), with adipocytes showing the more pronounced effect .
These findings suggest EGFL6 may be a molecular link between expanded adipose tissue and metabolic complications in obesity.
Yes, the obesity-associated increase in EGFL6 expression appears to be reversible. Analysis of paired adipose tissue samples before and after lifestyle-induced weight loss in well-characterized male individuals revealed:
Significant downregulation of EGFL6 expression in subcutaneous adipose tissue with increasing weight loss.
This downregulation occurred independently of changes in inflammatory markers (hs-CRP), leptin levels, or insulin resistance markers (HOMA-IR) .
This reversibility suggests that EGFL6 expression is dynamically regulated by adipose tissue status rather than being permanently altered in obesity, making it a potential indicator of successful weight management interventions.
For comprehensive investigation of EGFL6 in metabolic disorders, researchers should employ:
Tissue-specific expression analysis:
Correlation studies with metabolic parameters:
Functional studies:
In vitro adipocyte models to study EGFL6 effects on adipogenesis and metabolism
Co-culture systems to investigate paracrine effects between adipocytes and other cell types
These approaches allow for detailed characterization of EGFL6's role in obesity and associated metabolic dysfunction.
The development of clinically viable anti-EGFL6 antibodies requires a systematic approach:
CDR grafting optimization: When humanizing murine antibodies, researchers should generate multiple variant antibodies (e.g., 26 variants as described) with different VH/VL combinations to identify candidates with optimal binding while minimizing immunogenicity .
Affinity maturation techniques:
Identify key residues affecting antibody expression and binding affinity
Construct partially randomized NNK libraries (where N = A/C/G/T and K = G/T)
Screen approximately 90 clones per NNK library for antigen binding by ELISA
Design combinatory libraries with desirable mutations at 50% frequency
Screen approximately 400 clones for binding activity
This process has successfully generated humanized affinity-matured antibodies with KD values ranging from 150 pM to 2.67 nM compared to the parental humanized antibody (KD of 3.97 nM) .
Potential biomarkers for predicting response to anti-EGFL6 therapies include:
EGFL6 expression levels in tumor tissue, which can be assessed by immunohistochemistry, RNA sequencing, or quantitative PCR.
ERK pathway activation status, as EGFL6 activates ERK signaling and anti-EGFL6 antibodies inhibit EGFL6-mediated ERK phosphorylation .
Angiogenesis markers, since EGFL6 promotes endothelial cell proliferation and anti-EGFL6 therapy suppresses angiogenesis in tumor models .
Proliferation indices such as Ki67 expression, which is reduced in tumors treated with anti-EGFL6 antibodies .
Researchers should consider combining these markers to develop predictive signatures that could identify patients most likely to benefit from EGFL6-targeted interventions.
Despite significant advances, several critical knowledge gaps remain:
Receptor identification and signaling: The specific receptor(s) through which EGFL6 mediates its effects are not fully characterized. Understanding these receptor-ligand interactions could reveal additional therapeutic targets.
Cross-talk with other pathways: How EGFL6 signaling integrates with other growth factor pathways and inflammatory signaling networks requires further investigation.
Tissue-specific functions: EGFL6 is expressed in multiple tissues including placenta, adipose tissue, and hypothalamus . The physiological and pathological roles in these diverse tissues need clarification.
Broader disease relevance: While roles in ovarian cancer and obesity are emerging, EGFL6's potential involvement in other cancers and metabolic disorders remains to be fully explored.
Therapeutic resistance mechanisms: As with many targeted therapies, potential resistance mechanisms to anti-EGFL6 therapies need to be anticipated and addressed.
Addressing these knowledge gaps will require interdisciplinary approaches combining molecular biology, systems biology, and translational research methodologies.
Based on current knowledge, the most promising research directions include:
Dual-targeting therapeutic approaches: Combining anti-EGFL6 antibodies with therapies targeting complementary pathways may enhance efficacy against cancer.
EGFL6 as a biomarker: Further validation of EGFL6 as a prognostic and predictive biomarker in various cancers and metabolic disorders.
Mechanistic studies: Deeper investigation of EGFL6's molecular mechanisms, particularly receptor identification and signaling pathway interactions.
Expanded disease relevance: Exploration of EGFL6's role in other cancer types and metabolic conditions beyond those currently studied.
Clinical translation: Moving humanized affinity-matured antibodies toward first-in-human clinical trials, particularly in ovarian cancer where preclinical evidence is strongest .
These directions hold significant potential for advancing both basic understanding of EGFL6 biology and its clinical applications.
Several methodological innovations could significantly advance EGFL6 research:
Single-cell technologies: Application of single-cell RNA sequencing and proteomics to better understand cell-specific EGFL6 expression and response patterns.
Advanced in vivo models: Development of genetically engineered mouse models with tissue-specific EGFL6 expression or deletion to study physiological functions.
High-throughput functional screening: CRISPR-based functional genomic screens to identify genes that modify EGFL6 expression or signaling.
Structural biology approaches: Determination of EGFL6 protein structure and binding interfaces with receptors to guide rational design of inhibitors.
Biomarker development platforms: Integration of EGFL6 into liquid biopsy panels for non-invasive monitoring of cancer progression and treatment response.
These methodological advances would address current technical limitations and accelerate progress in understanding EGFL6 biology and developing therapeutic applications.
The EGFL6 gene encodes a protein that contains several distinct domains:
Recombinant human EGF-L6 protein is produced using human embryonic kidney cell (HEK293)-derived systems. The protein is purified to a high degree, with a purity greater than 95% as determined by SDS-PAGE . It is used in various research applications, including studies on cell adhesion and matrix assembly .