CD24 antibodies are monoclonal antibodies designed to bind CD24, a glycosylphosphatidylinositol (GPI)-anchored cell surface protein overexpressed in multiple cancers. CD24 interacts with Siglec-10 on macrophages and natural killer (NK) cells, transmitting a "don’t eat me" signal to suppress phagocytosis and immune activation .
CD24 antibodies exert anti-tumor effects through three primary mechanisms:
Immune Checkpoint Blockade
Macrophage Reprogramming
Direct Cytotoxicity
In MC38 colon cancer models, 10 mg/kg IMM47 eliminated tumors and conferred immunity to rechallenge .
ATG-031 showed dose-dependent phagocytosis enhancement in vitro (EC₅₀: 0.1–1 nM) .
Biomarker Development: CD24 expression varies widely; companion diagnostics are needed for patient stratification .
Combination Strategies: Synergy observed with anti-PD-1 therapy in preclinical models .
Resistance Mechanisms: Tumor cell CD24 downregulation observed in late-stage treatments .
CD24 is a small glycosylphosphatidylinositol (GPI)-anchored membrane sialoglycoprotein with a molecular weight of 35-45 kDa. Despite having a short protein core containing only 31-34 amino acids, CD24 exhibits extensive N-linked and O-linked glycosylation patterns . This glycoprotein was initially discovered in mice as a heat-stable antigen (HSA) . The extensive glycosylation of CD24 creates significant structural complexity beyond its small protein core, which contributes to its diverse functions in cellular processes and signaling pathways.
In healthy tissues, CD24 expression is primarily found on B cells from the pre-B to mature B cell stage, but notably absent on plasma cells. It is also expressed on mature granulocytes and various epithelial cell types . While CD24 is present in some normal tissues, its distribution in normal tissues is relatively limited compared to its extensive expression in tumor tissues . This differential expression pattern makes CD24 an attractive target for cancer therapy, though researchers must account for potential off-target effects in normal CD24-expressing tissues when designing therapeutic antibodies.
In normal immune cell development, CD24 serves as a costimulatory factor for T cells that regulates their homeostasis and proliferation. In B cells, CD24 is functionally involved in cell activation and differentiation . Additionally, CD24 in hematopoietic cells can bind to molecules exhibiting danger-associated molecular patterns (DAMPs) . CD24 mediates signal transduction and activates the mitogen-activated protein kinase pathway, which is involved in B- and T-cell development and apoptosis, cell binding, and other cellular processes.
CD24 is overexpressed in approximately 70% of human cancers, making it a common tumor-associated antigen . Research using immunohistochemistry (IHC) on tissue microarrays has demonstrated that CD24 is extensively expressed in a variety of tumor types, while its distribution in normal tissues remains limited . This widespread expression across multiple cancer types makes CD24 a promising target for broad-spectrum cancer immunotherapy development. Its high prevalence suggests that therapeutic approaches targeting CD24 could potentially benefit numerous cancer patients across different cancer types.
The CD24/Siglec-10 pathway represents a novel "don't eat me" signal in cancer. CD24 interacts with Siglec-10 expressed by tumor-associated macrophages, preventing the engulfment of cancer cells by these macrophages . This interaction functions as an innate immune checkpoint, allowing cancer cells to evade immune surveillance. Through this mechanism, overexpression of CD24 on tumor cells restricts the immune response and promotes tumor progression. Therapeutic antibodies blocking this interaction have exhibited potent preclinical and early clinical efficacy against both solid tumors and hematological malignancies .
CD24 has been identified as a marker for certain cancer stem cell populations. Specifically, CD24 is a marker for pancreatic and ovarian cancer stem cells, whereas breast cancer stem cells are notably negative for CD24 . This differential expression pattern highlights the complexity of using CD24 as a cancer stem cell marker across different malignancies. Understanding the relationship between CD24 and cancer stem cells is crucial for developing therapeutic strategies that can target these therapy-resistant cell populations.
For detecting CD24 expression in tissue samples, immunohistochemistry (IHC) on tissue microarrays is a widely used technique . Flow cytometry using fluorescently labeled anti-CD24 antibodies is another effective method, particularly for analyzing CD24 expression on cell surfaces . When performing IHC, researchers should be aware that glycosylation patterns of CD24 differ between normal and cancer cells, which may affect antibody binding. For optimal results, antibodies recognizing the protein backbone epitope rather than glycosylation-dependent epitopes might provide more consistent staining across different sample types.
Preclinical evaluation of anti-CD24 antibodies typically includes both in vitro and in vivo assessments. In vitro assessments include binding affinity measurements, phagocytosis assays, receptor blockage assays, ex vivo cytokine release, and antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) assays . For in vivo efficacy, researchers commonly use syngeneic models with human CD24-expressing tumors. Pharmacodynamic (PD) studies typically involve flow cytometry and single-cell RNA sequencing of tumor-infiltrating lymphocytes isolated from tumor samples following antibody administration . For toxicology and pharmacokinetic studies, non-human primates (NHPs) have been utilized with both the test antibody and surrogate antibodies specific to the primate CD24 .
When working with CD24 antibodies, essential validation steps include:
Confirming antibody specificity using both positive control cells (CD24-high expressing cancer cell lines) and negative control cells (CD24-knockout or naturally CD24-negative cell lines)
Verifying binding affinity to recombinant CD24 protein
Testing the ability to block CD24-Siglec-10 interaction
Assessing functional activity through phagocytosis assays
Confirming epitope specificity, especially distinguishing between normal and cancer-associated CD24 epitopes
Positive controls should include cell lines known to express high levels of CD24, while negative controls should include CD24-negative or CD24-knockout cell lines to confirm antibody specificity.
Anti-CD24 antibodies employ multiple mechanisms of action in cancer therapy:
Blocking the CD24-Siglec-10 "don't eat me" signal, which enhances macrophage-mediated phagocytosis of cancer cells
Inducing antibody-dependent cellular cytotoxicity (ADCC) with an EC50 reported as low as 0.27nM for some antibodies
Enhancing immune cell infiltration into tumor tissues, suggesting involvement of antibody-dependent cellular cytotoxicity (ADCC)
Different anti-CD24 antibody clones exhibit significant variations in epitope recognition and functional outcomes:
These differences highlight the importance of epitope selection in developing CD24 antibodies with optimal therapeutic efficacy and safety profiles.
Combination strategies with CD24 antibodies and standard chemotherapy have shown promising results. In the A549 lung cancer model, the addition of SWA11 monoclonal antibody to gemcitabine treatment strongly potentiated its anti-cancer efficacy . This synergistic effect suggests that CD24 targeting could be beneficial when combined with standard chemotherapy regimens. The mechanisms behind this enhanced efficacy may include:
CD24 antibodies altering the tumor microenvironment to make it more susceptible to chemotherapy
Increased immune cell infiltration enhancing the efficacy of cytotoxic agents
Changes in cytokine profiles that may sensitize cancer cells to chemotherapeutic drugs
Potential reduction of cancer stem cell populations that are typically resistant to conventional chemotherapy
When designing combination studies, researchers should carefully consider dosing schedules, potential toxicities, and mechanistic interactions between the antibody and chemotherapeutic agent.
Developing CD24 antibodies with improved tumor specificity faces several key challenges:
CD24 is expressed not only in various cancer cells but also in individual normal tissues such as the esophagus and thyroid
The glycosylation pattern of CD24 remains incompletely characterized, posing a significant challenge for antibody development
Identifying unique cancer-specific epitopes, like the NeoCD24 epitope that is shielded by glycans in normal cells but exposed in cancer cells
Balancing potent anti-tumor activity with minimal off-target effects
Addressing potential tumor resistance mechanisms
Future research should focus on detailed characterization of cancer-specific glycosylation patterns of CD24 to enable the design of antibodies targeting these unique epitopes.
Biomarker development for predicting response to anti-CD24 therapy remains an active area of research. Potential biomarkers to investigate include:
CD24 expression levels in tumor tissues (quantified by IHC or flow cytometry)
Siglec-10 expression on tumor-associated macrophages
Tumor microenvironment characteristics, including baseline immune cell infiltration patterns
Cancer stem cell markers in combination with CD24
Specific glycosylation patterns of CD24 in tumor samples
Integration of these biomarkers into clinical studies should follow a multi-faceted approach:
Include mandatory tumor biopsies before treatment and on-treatment when feasible
Develop companion diagnostic assays that specifically detect cancer-associated CD24 epitopes
Correlate biomarker levels with treatment outcomes to identify predictive thresholds
Explore combinations of biomarkers rather than single markers for improved prediction accuracy
The development of reliable biomarkers would facilitate personalized cancer treatment approaches with CD24-targeted therapies .